1
|
Yuasa-Kawada J, Kinoshita-Kawada M, Hiramoto M, Yamagishi S, Mishima T, Yasunaga S, Tsuboi Y, Hattori N, Wu JY. Neuronal guidance signaling in neurodegenerative diseases: Key regulators that function at neuron-glia and neuroimmune interfaces. Neural Regen Res 2026; 21:612-635. [PMID: 39995079 DOI: 10.4103/nrr.nrr-d-24-01330] [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: 10/31/2024] [Accepted: 01/27/2025] [Indexed: 02/26/2025] Open
Abstract
The nervous system processes a vast amount of information, performing computations that underlie perception, cognition, and behavior. During development, neuronal guidance genes, which encode extracellular cues, their receptors, and downstream signal transducers, organize neural wiring to generate the complex architecture of the nervous system. It is now evident that many of these neuroguidance cues and their receptors are active during development and are also expressed in the adult nervous system. This suggests that neuronal guidance pathways are critical not only for neural wiring but also for ongoing function and maintenance of the mature nervous system. Supporting this view, these pathways continue to regulate synaptic connectivity, plasticity, and remodeling, and overall brain homeostasis throughout adulthood. Genetic and transcriptomic analyses have further revealed many neuronal guidance genes to be associated with a wide range of neurodegenerative and neuropsychiatric disorders. Although the precise mechanisms by which aberrant neuronal guidance signaling drives the pathogenesis of these diseases remain to be clarified, emerging evidence points to several common themes, including dysfunction in neurons, microglia, astrocytes, and endothelial cells, along with dysregulation of neuron-microglia-astrocyte, neuroimmune, and neurovascular interactions. In this review, we explore recent advances in understanding the molecular and cellular mechanisms by which aberrant neuronal guidance signaling contributes to disease pathogenesis through altered cell-cell interactions. For instance, recent studies have unveiled two distinct semaphorin-plexin signaling pathways that affect microglial activation and neuroinflammation. We discuss the challenges ahead, along with the therapeutic potentials of targeting neuronal guidance pathways for treating neurodegenerative diseases. Particular focus is placed on how neuronal guidance mechanisms control neuron-glia and neuroimmune interactions and modulate microglial function under physiological and pathological conditions. Specifically, we examine the crosstalk between neuronal guidance signaling and TREM2, a master regulator of microglial function, in the context of pathogenic protein aggregates. It is well-established that age is a major risk factor for neurodegeneration. Future research should address how aging and neuronal guidance signaling interact to influence an individual's susceptibility to various late-onset neurological diseases and how the progression of these diseases could be therapeutically blocked by targeting neuronal guidance pathways.
Collapse
Affiliation(s)
| | | | | | - Satoru Yamagishi
- Department of Optical Neuroanatomy, Institute of Photonics Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takayasu Mishima
- Division of Neurology, Department of Internal Medicine, Sakura Medical Center, Toho University, Sakura, Japan
| | - Shin'ichiro Yasunaga
- Department of Biochemistry, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Yoshio Tsuboi
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Jane Y Wu
- Department of Neurology, Center for Genetic Medicine, Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
2
|
Thiesler H, Hildebrandt H. Polysialic acid-Siglec immune checkpoints of microglia and macrophages: Perspectives for therapeutic intervention. Neural Regen Res 2026; 21:661-662. [PMID: 39688555 DOI: 10.4103/nrr.nrr-d-24-01195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Affiliation(s)
- Hauke Thiesler
- Institute of Clinical Biochemistry, Hannover Medical School and Center for Systems Neuroscience Hannover, Hannover, Germany
| | | |
Collapse
|
3
|
Wu S, Chen J. Is age-related myelinodegenerative change an initial risk factor of neurodegenerative diseases? Neural Regen Res 2026; 21:648-658. [PMID: 40326982 DOI: 10.4103/nrr.nrr-d-24-00848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 11/25/2024] [Indexed: 05/07/2025] Open
Abstract
Myelination, the continuous ensheathment of neuronal axons, is a lifelong process in the nervous system that is essential for the precise, temporospatial conduction of action potentials between neurons. Myelin also provides intercellular metabolic support to axons. Even minor disruptions in the integrity of myelin can impair neural performance and increase susceptibility to neurological diseases. In fact, myelin degeneration is a well-known neuropathological condition that is associated with normal aging and several neurodegenerative diseases, including multiple sclerosis and Alzheimer's disease. In the central nervous system, compact myelin sheaths are formed by fully mature oligodendrocytes. However, the entire oligodendrocyte lineage is susceptible to changes in the biological microenvironment and other risk factors that arise as the brain ages. In addition to their well-known role in action potential propagation, oligodendrocytes also provide intercellular metabolic support to axons by transferring energy metabolites and delivering exosomes. Therefore, myelin degeneration in the aging central nervous system is a significant contributor to the development of neurodegenerative diseases. Interventions that mitigate age-related myelin degeneration can improve neurological function in aging individuals. In this review, we investigate the changes in myelin that are associated with aging and their underlying mechanisms. We also discuss recent advances in understanding how myelin degeneration in the aging brain contributes to neurodegenerative diseases and explore the factors that can prevent, slow down, or even reverse age-related myelin degeneration. Future research will enhance our understanding of how reducing age-related myelin degeneration can be used as a therapeutic target for delaying or preventing neurodegenerative diseases.
Collapse
Affiliation(s)
- Shuangchan Wu
- Sanhang Institute for Brain Science and Technology (SiBST), School of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi Province, China
- Shenzhen Research Institute of Northwestern Polytechnical University, Shenzhen, Guangdong Province, China
| | - Jun Chen
- Sanhang Institute for Brain Science and Technology (SiBST), School of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi Province, China
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| |
Collapse
|
4
|
Ren C, Chen M, Ren B, Zeng Y, Tan Q, Li Q, Zhang X, Fang Y, Zhou Y, Zhang W, Chen F, Bian B, Liu Y. Mesenchymal stem cell-derived small extracellular vesicles enhance the therapeutic effect of retinal progenitor cells in retinal degenerative disease rats. Neural Regen Res 2026; 21:821-832. [PMID: 39101643 DOI: 10.4103/nrr.nrr-d-23-02108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 06/08/2024] [Indexed: 08/06/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202602000-00050/figure1/v/2025-05-05T160104Z/r/image-tiff Our previous study demonstrated that combined transplantation of bone marrow mesenchymal stem cells and retinal progenitor cells in rats has therapeutic effects on retinal degeneration that are superior to transplantation of retinal progenitor cells alone. Bone marrow mesenchymal stem cells regulate and interact with various cells in the retinal microenvironment by secreting neurotrophic factors and extracellular vesicles. Small extracellular vesicles derived from bone marrow mesenchymal stem cells, which offer low immunogenicity, minimal tumorigenic risk, and ease of transportation, have been utilized in the treatment of various neurological diseases. These vesicles exhibit various activities, including anti-inflammatory actions, promotion of tissue repair, and immune regulation. Therefore, novel strategies using human retinal progenitor cells combined with bone marrow mesenchymal stem cell-derived small extracellular vesicles may represent an innovation in stem cell therapy for retinal degeneration. In this study, we developed such an approach utilizing retinal progenitor cells combined with bone marrow mesenchymal stem cell-derived small extracellular vesicles to treat retinal degeneration in Royal College of Surgeons rats, a genetic model of retinal degeneration. Our findings revealed that the combination of bone marrow mesenchymal stem cell-derived small extracellular vesicles and retinal progenitor cells significantly improved visual function in these rats. The addition of bone marrow mesenchymal stem cell-derived small extracellular vesicles as adjuvants to stem cell transplantation with retinal progenitor cells enhanced the survival, migration, and differentiation of the exogenous retinal progenitor cells. Concurrently, these small extracellular vesicles inhibited the activation of regional microglia, promoted the migration of transplanted retinal progenitor cells to the inner nuclear layer of the retina, and facilitated their differentiation into photoreceptors and bipolar cells. These findings suggest that bone marrow mesenchymal stem cell-derived small extracellular vesicles potentiate the therapeutic efficacy of retinal progenitor cells in retinal degeneration by promoting their survival and differentiation.
Collapse
Affiliation(s)
- Chunge Ren
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Min Chen
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Bangqi Ren
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Yuxiao Zeng
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Qiang Tan
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Qiyou Li
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Xue Zhang
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Yajie Fang
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Yixiao Zhou
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Weitao Zhang
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Fang Chen
- Department of Medical Technology, Chongqing Medical and Pharmaceutical College, Chongqing, China
| | - Baishijiao Bian
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
- Army 953 Hospital, Shigatse Branch of Xinqiao Hospital, Third Military Medical University (Army Medical University), Shigatse, Tibet Autonomous Region, China
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yong Liu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
- Jinfeng Laboratory, Chongqing, China
| |
Collapse
|
5
|
Esteban P, Letona-Gimenez S, Domingo MP, Morte E, Pellejero-Sagastizabal G, Del Mar Encabo M, Ramírez-Labrada A, Sanz-Pamplona R, Pardo J, Paño JR, Galvez EM. Combination of exhaled volatile organic compounds with serum biomarkers predicts respiratory infection severity. Pulmonology 2025; 31:2477911. [PMID: 40152323 DOI: 10.1080/25310429.2025.2477911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 03/06/2025] [Indexed: 03/29/2025] Open
Abstract
OBJECTIVE During respiratory infections, host-pathogen interaction alters metabolism, leading to changes in the composition of expired volatile organic compounds (VOCs) and soluble immunomodulators. This study aims to identify VOC and blood biomarker signatures to develop machine learning-based prognostic models capable of distinguishing infections with similar symptoms. METHODS Twenty-one VOCs and fifteen serum biomarkers were quantified in samples from 86 COVID-19 patients, 75 patients with non-COVID-19 respiratory infections, and 72 healthy donors. The populations were categorized into severity subgroups based on their oxygen support requirements. Descriptive and statistical analyses were conducted to assess group differentiation. Additionally, machine learning classifiers were developed to predict disease severity in both COVID-19 and non-COVID-19 patients. RESULTS VOC and biomarker profiles differed significantly among groups. Random Forest models demonstrated the best performance for severity prediction. The COVID-19 model achieved 93% accuracy, 100% sensitivity, and 89% specificity, identifying IL-6, IL-8, thrombomodulin, and toluene as key severity predictors. In non-COVID-19 patients, the model reached 89% accuracy, 100% sensitivity, and 67% specificity, with CXCL10 and methyl-isobutyl-ketone as key markers. CONCLUSION VOCs and serum biomarkers differentiated HD, COVID-19, and non-COVID-19 patients, and enabled the development of high-performance severity prediction models. While promising, these findings require validation in larger independent cohorts.
Collapse
Affiliation(s)
| | - Santiago Letona-Gimenez
- Servicio de Enfermedades Infecciosas, Hospital Clínico Universitario Lozano Blesa, Zaragoza, Spain
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
| | | | - Elena Morte
- Servicio de Enfermedades Infecciosas, Hospital Clínico Universitario Lozano Blesa, Zaragoza, Spain
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
- CIBERINFEC, ISCIII - CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Galadriel Pellejero-Sagastizabal
- Servicio de Enfermedades Infecciosas, Hospital Clínico Universitario Lozano Blesa, Zaragoza, Spain
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
| | | | - Ariel Ramírez-Labrada
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
- CIBERINFEC, ISCIII - CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Rebeca Sanz-Pamplona
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
- CIBERESP, ISCIII - CIBER de Epidemiologia y Salud Pública, Instituto de Salud Carlos III, Madrid, Spain
- Fundación Agencia Aragonesa para la Investigación y el Desarrollo (ARAID), Zaragoza, Spain
- Cancer Heterogeneity and Immunomics group, Hospital Clínico Universitario Lozano Blesa, Zaragoza, Spain
| | - Julián Pardo
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
- CIBERINFEC, ISCIII - CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
- Departamento de Microbiología, Pediatría, Radiología y Salud Pública, Área de Inmunología, Facultad de Medicina, Universidad de Zaragoza, Zaragoza, Spain
| | - José Ramón Paño
- Servicio de Enfermedades Infecciosas, Hospital Clínico Universitario Lozano Blesa, Zaragoza, Spain
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
- CIBERINFEC, ISCIII - CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Eva M Galvez
- Instituto de Carboquímica ICB-CSIC, Zaragoza, Spain
- CIBERINFEC, ISCIII - CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
6
|
Safi S, Krzykalla J, Hoffmann H, Benner A, Bischoff H, Eichhorn M, Kriegsmann M, Poschke I, Stögbauer F, Umansky L, Mogler C, Weichert W, Winter H, Beckhove P, Muley T. Low tumor interleukin-1β expression predicts a limited effect of adjuvant platinum-based chemotherapy for patients with completely resected lung adenocarcinoma: An identification and validation study. Pulmonology 2025; 31:2416803. [PMID: 38614857 DOI: 10.1016/j.pulmoe.2024.03.003] [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: 09/24/2023] [Revised: 02/27/2024] [Accepted: 03/23/2024] [Indexed: 04/15/2024] Open
Abstract
INTRODUCTION AND OBJECTIVES Adjuvant platinum-based chemotherapy for completely resected non-small cell lung cancer is associated with modest improvement in survival; nevertheless, no validated biomarker exists for predicting the benefit or harm of adjuvant platinum-based chemotherapy. MATERIALS AND METHODS We simultaneously measured 27 cytokines in operative tumor specimens from a discovery cohort (n = 97) by multiplex immunoassay; half of the patients received adjuvant platinum-based chemotherapy, and the other half were observed. We tested possible prognostic and predictive factors in multivariate Cox models for overall survival (OS) and relapse-free survival (RFS), and a tree-based method was applied to detect predictive factors with respect to RFS. The results were validated in an independent validation cohort (n = 93). RESULTS Fifty-two of 97 (54 %) patients in the discovery cohort and 50 of 93 (54 %) in the validation cohort received adjuvant chemotherapy; forty-four (85 %) patients in the discovery cohort and 37 (74 %) in the validation cohort received four cycles as planned. In patients with low IL-1β-expressing tumors, RFS and OS were worse after adjuvant chemotherapy than after observation. The limited effect of adjuvant chemotherapy for patients with low IL-1β-expressing tumors was confirmed in the validation cohort. Additionally, RFS and OS were prolonged by adjuvant chemotherapy only in patients with high IL-1β-expressing tumors in the validation cohort. CONCLUSIONS This study identified and validated low tumor IL-1β expression as a potential biomarker of a limited response to adjuvant platinum-based chemotherapy after complete resection of pulmonary adenocarcinoma. This finding has the potential to inform adjuvant treatment decisions.
Collapse
Affiliation(s)
- S Safi
- Division of Thoracic Surgery, University Hospital rechts der Isar, Technical University of Munich, Munich, Germany
| | - J Krzykalla
- Division of Biostatistics, German Cancer Research Center, Heidelberg, Germany
| | - H Hoffmann
- Division of Thoracic Surgery, University Hospital rechts der Isar, Technical University of Munich, Munich, Germany
| | - A Benner
- Division of Biostatistics, German Cancer Research Center, Heidelberg, Germany
| | - H Bischoff
- Department of Thoracic Oncology, Thoraxklinik, University of Heidelberg, Heidelberg, Germany
| | - M Eichhorn
- Department of Thoracic Surgery, Thoraxklinik, Heidelberg University Hospital, Heidelberg, Germany
| | - M Kriegsmann
- Institute of Pathology, Heidelberg University, Heidelberg, Germany
| | - I Poschke
- Immune Monitoring Unit, National Center for Tumor Diseases, Heidelberg, Germany
| | - F Stögbauer
- Institute of Pathology, Technical University of Munich, Munich, Germany
| | - L Umansky
- Immune Monitoring Unit, National Center for Tumor Diseases, Heidelberg, Germany
- Skin Cancer Unit, German Cancer Research Center, Heidelberg, Germany
| | - C Mogler
- Institute of Pathology, Technical University of Munich, Munich, Germany
| | - W Weichert
- Institute of Pathology, Technical University of Munich, Munich, Germany
| | - H Winter
- Department of Thoracic Surgery, Thoraxklinik, Heidelberg University Hospital, Heidelberg, Germany
| | - P Beckhove
- Regensburg Center for Interventional Immunology, University of Regensburg, Regensburg, Germany
| | - T Muley
- Translational Research Unit, Thoraxklinik, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
7
|
Münz C, Campbell GR, Esclatine A, Faure M, Labonte P, Lussignol M, Orvedahl A, Altan-Bonnet N, Bartenschlager R, Beale R, Cirone M, Espert L, Jung J, Leib D, Reggiori F, Sanyal S, Spector SA, Thiel V, Viret C, Wei Y, Wileman T, Wodrich H. Autophagy machinery as exploited by viruses. AUTOPHAGY REPORTS 2025; 4:27694127.2025.2464986. [PMID: 40201908 PMCID: PMC11921968 DOI: 10.1080/27694127.2025.2464986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/17/2025] [Accepted: 01/27/2025] [Indexed: 04/10/2025]
Abstract
Viruses adapt and modulate cellular pathways to allow their replication in host cells. The catabolic pathway of macroautophagy, for simplicity referred to as autophagy, is no exception. In this review, we discuss anti-viral functions of both autophagy and select components of the autophagy machinery, and how viruses have evaded them. Some viruses use the membrane remodeling ability of the autophagy machinery to build their replication compartments in the cytosol or efficiently egress from cells in a non-lytic fashion. Some of the autophagy machinery components and their remodeled membranes can even be found in viral particles as envelopes or single membranes around virus packages that protect them during spreading and transmission. Therefore, studies on autophagy regulation by viral infections can reveal functions of the autophagy machinery beyond lysosomal degradation of cytosolic constituents. Furthermore, they can also pinpoint molecular interactions with which the autophagy machinery can most efficiently be manipulated, and this may be relevant to develop effective disease treatments based on autophagy modulation.
Collapse
Affiliation(s)
- Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich Switzerland
| | - Grant R Campbell
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of SD, Vermillion, SD, USA
| | - Audrey Esclatine
- Université Paris-Saclay, CEA, CNRS, 10 Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Mathias Faure
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Universite Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007 Lyon, France
| | - Patrick Labonte
- eINRS-Centre Armand-Frappier Santé Biotechnologie, Laval, Canada
| | - Marion Lussignol
- Université Paris-Saclay, CEA, CNRS, 10 Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Anthony Orvedahl
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO, USA
| | - Nihal Altan-Bonnet
- Laboratory of Host-Pathogen Dynamics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ralf Bartenschlager
- Heidelberg University, Medical Faculty Heidelberg, Department of Infectious Diseases, Molecular Virology, Heidelberg, Germany
- German Cancer Research Center (DKFZ), Division Virus-Associated Carcinogenesis, Heidelberg, Germany
- German Centre for Infection Research, Heidelberg partner site, Heidelberg, Germany
| | - Rupert Beale
- Cell Biology of Infection Laboratory, The Francis Crick Institute, London, UK
- Division of Medicine, University College London, London, UK
| | - Mara Cirone
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Lucile Espert
- University of Montpellier, Montpellier, France
- CNRS, Institut de Recherche enInfectiologie deMontpellier (IRIM), Montpellier, France
| | - Jae Jung
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - David Leib
- Guarini School of Graduate and Advanced Studies at Dartmouth, Hanover, NH, USA
| | - Fulvio Reggiori
- Department of Biomedicine, Aarhus University, Ole Worms Allé 4, Aarhus C, Denmark
| | - Sumana Sanyal
- Sir William Dunn School of Pathology, South Parks Road, University of Oxford, Oxford, UK
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Stephen A. Spector
- Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Rady Children’s Hospital, San Diego, CA, USA
| | - Volker Thiel
- Institute of Virology and Immunology, Bern and Mittelhäusern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland, and Multidisciplinary Center for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Christophe Viret
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Universite Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007 Lyon, France
| | - Yu Wei
- Institut Pasteur-Theravectys Joint Laboratory, Department of Virology, Institut Pasteur, Université Paris Cité, Paris, France
| | - Thomas Wileman
- Norwich Medical School, University of East Anglia
- Quadram Institute Bioscience, Norwich Research Park, Norfolk, UK
| | - Harald Wodrich
- sLaboratoire de Microbiologie Fondamentale et Pathogénicité, MFP CNRS UMR, Université de Bordeaux, Bordeaux, France
| |
Collapse
|
8
|
Islam S, Chauhan VM, Pantazes RJ. Analysis of how antigen mutations disrupt antibody binding interactions toward enabling rapid and reliable antibody repurposing. MAbs 2025; 17:2440586. [PMID: 39690439 DOI: 10.1080/19420862.2024.2440586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/19/2024] Open
Abstract
Antibody repurposing is the process of changing a known antibody so that it binds to a mutated antigen. One of the findings to emerge from the Coronavirus Disease 2019 (COVID-19) pandemic was that it was possible to repurpose neutralizing antibodies for Severe Acute Respiratory Syndrome, a related disease, to work for COVID-19. Thus, antibody repurposing is a possible pathway to prepare for and respond to future pandemics, as well as personalizing cancer therapies. For antibodies to be successfully repurposed, it is necessary to know both how antigen mutations disrupt their binding and how they should be mutated to recover binding, with this work describing an analysis to address the first of these topics. Every possible antigen point mutation in the interface of 246 antibody-protein complexes were analyzed using the Rosetta molecular mechanics force field. The results highlight a number of features of how antigen mutations affect antibody binding, including the effects of mutating critical hotspot residues versus other positions, how many mutations are necessary to be likely to disrupt binding, the prevalence of indirect effects of mutations on binding, and the relative importance of changing attractive versus repulsive energies. These data are expected to be useful in guiding future antibody repurposing experiments.
Collapse
Affiliation(s)
- Sumaiya Islam
- Department of Chemical Engineering, Auburn University, Auburn, AL, USA
| | - Varun M Chauhan
- Department of Chemical Engineering, Auburn University, Auburn, AL, USA
| | - Robert J Pantazes
- Department of Chemical Engineering, Auburn University, Auburn, AL, USA
| |
Collapse
|
9
|
Chen J, Tarantino G, Severgnini M, Baginska J, Giobbie-Hurder A, Weirather JL, Manos M, Russell JD, Pfaff KL, Rodig SJ, Huang AY, Brennick R, Nazzaro M, Hathaway E, Holovatska M, Manuszak C, Ranasinghe S, Liu D, Hodi FS. Circulating cytokine associations with clinical outcomes in melanoma patients treated with combination nivolumab plus ipilimumab. Oncoimmunology 2025; 14:2432723. [PMID: 39699928 DOI: 10.1080/2162402x.2024.2432723] [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: 10/18/2024] [Accepted: 11/18/2024] [Indexed: 12/20/2024] Open
Abstract
Nivolumab plus ipilimumab (aCTLA-4/aPD-1) combination therapy has significantly improved clinical outcomes in patients with metastatic melanoma, with 50%-60% of patients responding to treatment, but predictors of response are poorly characterized. We hypothesized that circulating cytokines and peripheral white blood cells may predict response to therapy and evaluated 15 cytokines and complete blood counts (CBC with differentials) from 89 patients with advanced melanoma treated with combination therapy from three points in time: pre-treatment, one month and approximately three months after starting therapy. Clinical endpoints evaluated included durable clinical benefit (DCB), progression-free survival (PFS), and overall survival (OS). A parsimonious predictive model was developed to identify cytokines predictors of response to combination therapy. In this study, we found that pre-treatment, patients with DCB had higher IL-23, lower CXCL6, and lower IL-10 levels. Lower NLR one month after starting therapy predicted better PFS and OS, primarily driven by an increase in absolute lymphocytes. A multivariate model demonstrated that baseline CXCL6, IL-10, IL-23 were independent predictors of therapy response, and the combined model has reached an area under the curve (AUC) of 0.79 in prediction of response to combination therapy. Our study identified baseline CXCL6, IL-23, and IL-10 as predictors of response to aCTLA4/aPD1 combination therapy among patients with metastatic melanoma. This study also provides a framework for identifying patients who are likely to respond to combination ICB, as well as a subset of patients with high risk of developing resistance and are thus in need of alternative therapeutic options, such as clinical trials.
Collapse
Affiliation(s)
- Jiajia Chen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Giuseppe Tarantino
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Mariano Severgnini
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Joanna Baginska
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Anita Giobbie-Hurder
- Division of Biostatistics, Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jason L Weirather
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Michael Manos
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Janice D Russell
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kathleen L Pfaff
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Scott J Rodig
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Amy Y Huang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Ryan Brennick
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Matthew Nazzaro
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Emma Hathaway
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Marta Holovatska
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Claire Manuszak
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Srinika Ranasinghe
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - David Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - F Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
10
|
Guimarães JG, de Campos GY, Machado MP, Oliveira Brito PKM, dos Reis TF, Goldman GH, Bonini Palma PV, de Campos Fraga-Silva TF, Cavallin DCU, Venturini J, da Silva TA. A novel mannan-specific chimeric antigen receptor M-CAR redirects T cells to interact with Candida spp. hyphae and Rhizopus oryzae spores. Bioengineered 2025; 16:2458786. [PMID: 39891522 PMCID: PMC11792852 DOI: 10.1080/21655979.2025.2458786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 07/24/2024] [Accepted: 08/28/2024] [Indexed: 02/03/2025] Open
Abstract
Invasive fungal infections (IFIs) are responsible for elevated rates of morbidity and mortality, causing around of 1.5 million deaths annually worldwide. One of the main causative agents of IFIs is Candida albicans, and non-albicans Candida species have emerged as a spreading global public health concernment. Furthermore, COVID-19 has contributed to a boost in the incidence of IFIs, such as mucormycosis, in which Rhizopus oryzae is the most prevalent causative agent. The effector host immune response against IFIs depends on the activity of T cells, which are susceptible to the regulatory effects triggered by fungal virulence factors. The fungal cell wall plays a crucial role as a virulence factor, and its remodeling compromises the development of a specific T-cell response. The redirection of Jurkat T cells to target Candida spp. by recognizing targets expressed on the fungal cell wall can be facilitated using chimeric antigen receptor (CAR) technology. This study generated an M-CAR that contains an scFv with specificity to α-1,6 mannose backbone of fungal mannan, and the expression of M-CAR on the surface of modified Jurkat cells triggered a strong activation against Candida albicans (hyphae form), Candida tropicalis (hyphae form), Candida parapsilosis (pseudohyphal form), and Candida glabrata (yeast form). Moreover, M-CAR Jurkat cells recognized Rhizopus oryzae spores, which induced high expression of cell activation markers. Thus, a novel Mannan-specific CAR enabled strong signal transduction in modified Jurkat cells in the presence of Candida spp. or R. oryzae.
Collapse
Affiliation(s)
- Júlia Garcia Guimarães
- Department of Cellular and Molecular Biology, Ribeirao Preto Medical School, University of São Paulo, São Paulo, Brazil
- Department of Clinical Analysis, School of Pharmaceutical Sciences in Araraquara, Sao Paulo State University, São Paulo, Brazil
| | - Gabriela Yamazaki de Campos
- Department of Cellular and Molecular Biology, Ribeirao Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Michele Procópio Machado
- Department of Cellular and Molecular Biology, Ribeirao Preto Medical School, University of São Paulo, São Paulo, Brazil
| | | | | | | | - Patricia Vianna Bonini Palma
- Center for Cell-Based Therapy, Regional Blood Center of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | | | | | - James Venturini
- Faculty of Medicine, Federal University of Mato Grosso do Sul, Campo Grande, Brazil
| | - Thiago Aparecido da Silva
- Department of Cellular and Molecular Biology, Ribeirao Preto Medical School, University of São Paulo, São Paulo, Brazil
- Department of Clinical Analysis, School of Pharmaceutical Sciences in Araraquara, Sao Paulo State University, São Paulo, Brazil
| |
Collapse
|
11
|
Xu H, Cao L, Chen Y, Zhou C, Xu J, Zhang Z, Li X, Liu L, Lu J. Single-cell RNA sequencing reveals the heterogeneity and interactions of immune cells and Müller glia during zebrafish retina regeneration. Neural Regen Res 2025; 20:3635-3648. [PMID: 38934409 PMCID: PMC11974639 DOI: 10.4103/nrr.nrr-d-23-02083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 04/17/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202512000-00031/figure1/v/2025-01-31T122243Z/r/image-tiff Inflammation plays a crucial role in the regeneration of fish and avian retinas. However, how inflammation regulates Müller glia (MG) reprogramming remains unclear. Here, we used single-cell RNA sequencing to investigate the cell heterogeneity and interactions of MG and immune cells in the regenerating zebrafish retina. We first showed that two types of quiescent MG (resting MG1 and MG2) reside in the uninjured retina. Following retinal injury, resting MG1 transitioned into an activated state expressing known reprogramming genes, while resting MG2 gave rise to rod progenitors. We further showed that retinal microglia can be categorized into three subtypes (microglia-1, microglia-2, and proliferative) and pseudotime analysis demonstrated dynamic changes in microglial status following retinal injury. Analysis of cell-cell interactions indicated extensive crosstalk between immune cells and MG, with many interactions shared among different immune cell types. Finally, we showed that inflammation activated Jak1-Stat3 signaling in MG, promoting their transition from a resting to an activated state. Our study reveals the cell heterogeneity and crosstalk of immune cells and MG in zebrafish retinal repair, and may provide valuable insights into future mammalian retina regeneration.
Collapse
Affiliation(s)
- Hui Xu
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
- Key Lab of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, China
| | - Lining Cao
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yuxi Chen
- Key Lab of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, China
| | - Cuiping Zhou
- Key Lab of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, China
| | - Jie Xu
- Key Lab of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, China
| | - Zhuolin Zhang
- Key Lab of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, China
| | - Xiangyu Li
- Key Lab of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, China
| | - Lihua Liu
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Jianfeng Lu
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| |
Collapse
|
12
|
Naspolini NF, Schüroff PA, Vanzele PAR, Pereira-Santos D, Valim TA, Bonham KS, Fujita A, Passos-Bueno MR, Beltrão-Braga PCB, Carvalho ACPLF, Klepac-Ceraj V, Polanczyk GV, Campos AC, Taddei CR. Exclusive breastfeeding is associated with the gut microbiome maturation in infants according to delivery mode. Gut Microbes 2025; 17:2493900. [PMID: 40237336 PMCID: PMC12005435 DOI: 10.1080/19490976.2025.2493900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 01/29/2025] [Accepted: 04/08/2025] [Indexed: 04/18/2025] Open
Abstract
Exclusive breastfeeding (EBF) plays a crucial role in infant gut microbiome assembly and development. However, few studies have investigated the effects of EBF in restoring a perturbed microbiome. In this study, we applied whole metagenomic sequencing to assess the gut microbiome assembly in 525 Brazilian infants from 3 to 9 months of age of the Germina Cohort, demonstrating the early determinants of microbial taxonomy and function modulation. Our analysis shows that EBF alters the relative abundance of genes related to the microbiome taxonomy and function, with effects varying by delivery mode. EBF alters the pattern of carbohydrates, lipid metabolism, and cell structure pathways depending on the delivery mode. The microbiome age is closer to chronological infant age in EBF than in non-EBF infants, meaning a lower microbiome maturation index (MMI). Using a complementary machine learning approach, we show that Escherichia coli, Ruminococcus gnavus, and Clostridium neonatale, as well as vitamin K and o-antigen pathways contribute strongly to EBF prediction. Moreover, EBF influences the microbiome maturation in early life, toward a microbiome age more similar to the chronological infant's age.
Collapse
Affiliation(s)
| | - Paulo A. Schüroff
- School of Arts, Sciences and Humanity, University of Sao Paulo, Sao Paulo, Brazil
| | - Pedro A. R. Vanzele
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Davi Pereira-Santos
- Department of Applied Mathematics and Statistics, Institute of Mathematics and Computer Sciences, University of Sao Paulo, Sao Carlos, Brazil
- Departamento Acadêmico de Computação, Universidade Tecnológica Federal do Paraná (UTFPR), Câmpus Medianeira, Medianeira, Brazil
| | - Tamires Amabili Valim
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | - Kevin S. Bonham
- Department of Biological Sciences, Wellesley College, Wellesley, MA, USA
| | - André Fujita
- Division of Network AI Statistics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
- Department of Computer Science, Institute of Mathematics and Statistics, University of Sao Paulo, Sao Paulo, Brazil
| | - Maria Rita Passos-Bueno
- Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Patricia C. B. Beltrão-Braga
- Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
- Laboratory of Disease Modeling, Institut Pasteur de Sao Paulo, Sao Paulo, Brazil
| | - André C. P. L. F. Carvalho
- Department of Applied Mathematics and Statistics, Institute of Mathematics and Computer Sciences, University of Sao Paulo, Sao Carlos, Brazil
| | - Vanja Klepac-Ceraj
- Department of Biological Sciences, Wellesley College, Wellesley, MA, USA
| | - Guilherme V. Polanczyk
- Department of Psychiatry, Faculdade de Medicina FMUSP, University of Sao Paulo, Sao Paulo, Brazil
| | - Alline C. Campos
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | - Carla R. Taddei
- Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
- Division of Clinical Laboratory, University Hospital - University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
13
|
Qiao Y, Xie D, Li Z, Cao S, Zhao D. Global research trends on biomarkers for cancer immunotherapy: Visualization and bibliometric analysis. Hum Vaccin Immunother 2025; 21:2435598. [PMID: 39773010 PMCID: PMC11730411 DOI: 10.1080/21645515.2024.2435598] [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: 09/05/2024] [Revised: 11/08/2024] [Accepted: 11/21/2024] [Indexed: 01/11/2025] Open
Abstract
The global burden of cancer continues to grow, posing a significant public health challenge. Although cancer immunotherapy has shown significant efficacy, the response rate is not high. Therefore, the objective of our research was to identify the latest research trends and hotspots on biomarkers from 1993 to 2023. Data were collected from the database Web of Science core collection. Bibliometric analysis and visualization were conducted with CiteSpace(6.3.1), VOSviewer (v1.6.20), R-bibliometrix(v4.3.3), and Microsoft Excel(2019). A total of 2686 literatures were retrieved. The sheer annual volume of publications has shown a rapid upward trend since 2015. The United States has generated the most publications and Harvard University ranked as a leading institution. The global biomarker research on immune checkpoint inhibitors (ICIs) revealed regional differences and in-depth explorations should be promoted in developing countries. Although China has become the second largest country in terms of publication, the average citation per paper and the total link strength were both lower than the other countries. The research on biomarkers mainly concentrated upon the following aspects: PD-1/PD-L1, CTLA-4, gene expression, adverse events, total mutational burden (TMB), body mass index (BMI), gut microbiota, cd8(+)/cd4(+) t-cells, and blood-related biomarkers such as lactate dehydrogenase (LDH), neutrophil-lymphocyte ratio (NLR), cytokines. Furthermore, "artificial intelligence" and "machine learning" have become the most important research hotspot over the last 2 y, which will help us to identify useful biomarkers from complex big data and provide a basis for precise medicine for malignant tumors.
Collapse
Affiliation(s)
- Yuan Qiao
- Department of Clinical Pharmacy, Yan’an University Affiliated Hospital, Yan’an, Shaanxi, China
| | - Dong Xie
- Department of Pharmacy, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhengxiang Li
- Department of Pharmacy, Tianjin Medical University General Hospital, Tianjin, China
| | - Shaohua Cao
- Department of Clinical Pharmacy, Yan’an University Affiliated Hospital, Yan’an, Shaanxi, China
| | - Dong Zhao
- Department of Clinical Laboratory, Yan’an University Affiliated Hospital, Yan’an, Shaanxi, China
| |
Collapse
|
14
|
Lin HY, Ramos RB, Crawford DR. Oxidation of human mitochondrial RNA strongly potentiates immunostimulation in an interferon-associated manner. Redox Rep 2025; 30:2491845. [PMID: 40247667 PMCID: PMC12010657 DOI: 10.1080/13510002.2025.2491845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2025] Open
Abstract
Inflammation is associated with a wide range of medical conditions, most leading causes of death, and high healthcare costs. It can thus benefit from new insights. Here we extended previous studies and found that oxidation of human native mtRNA to 'mitoxRNA' strongly potentiated IFNβ and TNFα immunostimulation in human cells, and that this newly identified type 1 interferon potentiation was transcriptional. This potentiation was significantly greater than with mtDNA oxidation, and t-butylhydroperoxide (tBHP) oxidation of RNA was more proinflammatory than hydrogen peroxide (HP). mtRNA triggered a modest increase in apoptosis that was not potentiated by oxidation, and mtDNA triggered a much greater increase. For native mtRNA, we found that chloroquine-inhibitable endosomes and MDA5 are key signaling pathways for IFNβ and TNFα production. For mitoxRNAs, RNAseq revealed a major increase in both tBHP- and HP-mitoxRNA modulated genes compared with native mtRNA. This increase was very prominent for interferon-related genes, identifying them as important mediators of this powerful oxidation effect. Moderately different gene modulations and KEGG pathways were observed for tBHP- versus HP-mitoxRNAs. These studies reveal the profound effect that mitochondrial RNA oxidation has on immunostimulation, providing new insights into DAMP inflammation and identifying potential therapeutic targets to minimize DAMP mtRNA/mitoxRNA-mediated inflammation.
Collapse
Affiliation(s)
- Hung-Yun Lin
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
- Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - Ramon B. Ramos
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Dana R. Crawford
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA
| |
Collapse
|
15
|
Binte Hanafi Z, Mei Y, Teo HY, Zhu Y, Yong Lionel CC, Chiu JW, Lu J, Liu H. Calpain 2 regulates IL-1α secretion and inhibits tumor development via modulating calpain 1 expression in the tumor microenvironment. Oncoimmunology 2025; 14:2451444. [PMID: 39803956 PMCID: PMC11730618 DOI: 10.1080/2162402x.2025.2451444] [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: 09/21/2024] [Revised: 12/26/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025] Open
Abstract
Tumor-promoting inflammation significantly impacts cancer progression, and targeting inflammatory cytokines has emerged as a promising therapeutic approach in clinical trials. Interleukin (IL)-1α, a member of the IL-1 cytokine family, plays a crucial role in both inflammation and carcinogenesis. How IL-1α is secreted in the tumor microenvironment has been poorly understood, and we previously showed that calpain 1 cleaves pro-IL-1α for mature IL-1α secretion, which exacerbates hepatocellular carcinoma by recruiting myeloid-derived suppressor cells. In this study, we report that calpain 2 also modulates IL-1α secretion. Notably, a deficiency in calpain 2 resulted in enhanced hepatocellular carcinoma development within an IL-1α-enriched tumor microenvironment. Further investigations revealed that calpain 2 deficiency increased calpain 1 expression, implying a compensatory mechanism between the two calpains. Mechanistically, calpain 2 deficiency led to increased expression of FoxO3, which is a forkhead transcription factor that promotes calpain 1 expression. Collectively, these results suggest that calpain 2 modulates calpain 1 expression, and therefore IL-1α secretion through the induction of FoxO3, offering novel potential therapeutic targets for cancer treatment.
Collapse
Affiliation(s)
- Zuhairah Binte Hanafi
- Immunology Programme, Life Sciences Institute; Centre for Life Sciences, National University of Singapore, Singapore, Singapore
- Immunology Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yu Mei
- Immunology Programme, Life Sciences Institute; Centre for Life Sciences, National University of Singapore, Singapore, Singapore
- Immunology Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Huey Yee Teo
- Immunology Programme, Life Sciences Institute; Centre for Life Sciences, National University of Singapore, Singapore, Singapore
- Immunology Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ying Zhu
- Immunology Programme, Life Sciences Institute; Centre for Life Sciences, National University of Singapore, Singapore, Singapore
- Immunology Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chew Chin Yong Lionel
- Immunology Programme, Life Sciences Institute; Centre for Life Sciences, National University of Singapore, Singapore, Singapore
- Immunology Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jing Wen Chiu
- Immunology Programme, Life Sciences Institute; Centre for Life Sciences, National University of Singapore, Singapore, Singapore
- Immunology Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jinhua Lu
- Immunology Programme, Life Sciences Institute; Centre for Life Sciences, National University of Singapore, Singapore, Singapore
- Immunology Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Haiyan Liu
- Immunology Programme, Life Sciences Institute; Centre for Life Sciences, National University of Singapore, Singapore, Singapore
- Immunology Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
16
|
Crescioli S, Jatiani S, Moise L. With great power, comes great responsibility: the importance of broadly measuring Fc-mediated effector function early in the antibody development process. MAbs 2025; 17:2453515. [PMID: 39819511 PMCID: PMC11810086 DOI: 10.1080/19420862.2025.2453515] [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: 10/23/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 01/19/2025] Open
Abstract
The field of antibody therapeutics is rapidly growing, with over 210 antibodies currently approved or in regulatory review and ~ 1,250 antibodies in clinical development. Antibodies are highly versatile molecules that, with strategic design of their antigen-binding domain (Fab) and the domain responsible for mediating effector functions (Fc), can be used in a wide range of therapeutic indications. Building on many years of progress, the biopharmaceutical industry is now advancing innovative research and development by exploring new targets and new formats and using antibody engineering to fine-tune functions tailored to specific disease requirements. In addition to considering the target and the disease context, however, the unique features of each therapeutic antibody trigger a diverse set of Fc-mediated effector functions. To avoid unexpected results on safety and efficacy outcomes during the later stages of the development process, it is crucial to measure the impact of antibody design on Fc-mediated effector function early in the antibody development process. Given the breadth of effector functions antibodies can deploy and the close interplay between the antibody Fab and Fc functional domains, it is important to conduct a comprehensive evaluation of Fc-mediated functions using an array of antigen-specific biophysical and cell-mediated functional assays. Here, we review antibody and Fc receptor properties that influence Fc effector functions and discuss their implications on development of safe and efficacious antibody therapeutics.
Collapse
|
17
|
Ruple HK, Haasis E, Bettenburg A, Maier C, Fritz C, Schüle L, Löcker S, Soltow Y, Schintgen L, Schmidt NS, Schneider C, Lorentz A, Fricke WF. The gut microbiota predicts and time-restricted feeding delays experimental colitis. Gut Microbes 2025; 17:2453019. [PMID: 39843997 PMCID: PMC11758946 DOI: 10.1080/19490976.2025.2453019] [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: 11/14/2024] [Revised: 12/20/2024] [Accepted: 01/07/2025] [Indexed: 01/24/2025] Open
Abstract
The etiology of inflammatory bowel disease (IBD) remains unclear, treatment options unsatisfactory and disease development difficult to predict for individual patients. Dysbiosis of the gastrointestinal microbiota and disruption of the biological clock have been implicated and studied as diagnostic and therapeutic targets. Here, we examine the relationship of IBD to biological clock and gut microbiota by using the IL-10 deficient (IL-10-/-) mouse model for microbiota-dependent spontaneous colitis in combination with altered (4 h/4 h) light/dark cycles to disrupt and time-restricted feeding (TRF) to restore circadian rhythmicity. We show that while altered light/dark cycles disrupted the intestinal clock in wild type (WT) mice, IL-10-/- mice were characterized by altered microbiota composition, impaired intestinal clock, and microbiota rhythmicity irrespective of external clock disruption, which had no consistent colitis-promoting effect on IL-10-/- mice. TRF delayed colitis onset reduced the expression of inflammatory markers and increased the expression of clock genes in the intestine, and increased gut microbiota rhythmicity in IL-10-/- mice. Compositional changes and reduced rhythmicity of the fecal microbiota preceded colitis and could predict colitis symptoms for individual IL-10-/- mice across different experiments. Our findings provide perspectives for new diagnostic and TRF-based, therapeutic applications in IBD that should be further explored.
Collapse
Affiliation(s)
- Hannah K. Ruple
- Department of Microbiome Research and Applied Bioinformatics, Institute for Nutritional Sciences, University of Hohenheim, Stuttgart, Germany
| | - Eva Haasis
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - Anna Bettenburg
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - Carina Maier
- Department of Microbiome Research and Applied Bioinformatics, Institute for Nutritional Sciences, University of Hohenheim, Stuttgart, Germany
| | - Carolin Fritz
- Department of Microbiome Research and Applied Bioinformatics, Institute for Nutritional Sciences, University of Hohenheim, Stuttgart, Germany
| | - Laura Schüle
- Department of Microbiome Research and Applied Bioinformatics, Institute for Nutritional Sciences, University of Hohenheim, Stuttgart, Germany
| | - Sarah Löcker
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - Yvonne Soltow
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - Lynn Schintgen
- Department of Microbiome Research and Applied Bioinformatics, Institute for Nutritional Sciences, University of Hohenheim, Stuttgart, Germany
| | - Nina S. Schmidt
- Department of Microbiome Research and Applied Bioinformatics, Institute for Nutritional Sciences, University of Hohenheim, Stuttgart, Germany
| | - Celine Schneider
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - Axel Lorentz
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - W. Florian Fricke
- Department of Microbiome Research and Applied Bioinformatics, Institute for Nutritional Sciences, University of Hohenheim, Stuttgart, Germany
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
18
|
Li Z, Jiang S, Liu W, Yang X, Liu F, Li X, Li J, Yu M, Wei Z, Wang B, Qian D. A promising endeavor against human cytomegalovirus: Predominant epitopes-based recombinant subunit vaccine RH EcIE1/pp65/pp150. Virulence 2025; 16:2497903. [PMID: 40277436 PMCID: PMC12064061 DOI: 10.1080/21505594.2025.2497903] [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: 01/22/2024] [Revised: 12/23/2024] [Accepted: 04/21/2025] [Indexed: 04/26/2025] Open
Abstract
Human cytomegalovirus (HCMV) is widespread in the population, typically remaining latent. However, it can cause severe morbidity and mortality in transplant patients and immunodeficient individuals. Currently, there is no approved vaccine against HCMV. This study used immunoinformatics methods to predict the predominant T and B-cell epitopes of three key HCMV proteins, including phosphoprotein 65 (pp65), pp150, and immediate-early protein 1 (IE1). Subsequently, we synthesized a recombinant subunit vaccine (RHEcIE1/pp65/pp150) from Escherichia coli, comprising RHEc-1 and RHEc-2. We observed that the RHEcIE1/pp65/pp150 vaccine exhibited high safety and immunogenicity in mice, enhancing a significant upregulation of CD80, CD86, CD40, and MHCII on dendritic cells and macrophages. Additionally, the vaccine activated innate immune responses through the NF-κB signalling pathway, triggering CD4+ and CD8+T cells to secrete tumour necrosis factor (TNF)-α, interferon (IFN)-γ, and interleukin (IL)-2, directing the T-cell response towards Th1. Moreover, it stimulated CD4+T cells to secrete IL-4, IL-6, and IL-10, promoting B-cell immunity. Furthermore, the RHEcIE1/pp65/pp150 vaccine induced the formation of abundant memory cells and high levels of neutralizing antibody titres, conducive to providing long-lasting protection. Taken together, the RHEcIE1/pp65/pp150 vaccine is a promising endeavour against HCMV, and these findings contribute valuable insights to the development of HCMV vaccine candidates.
Collapse
MESH Headings
- Cytomegalovirus Vaccines/immunology
- Cytomegalovirus Vaccines/genetics
- Cytomegalovirus Vaccines/administration & dosage
- Animals
- Vaccines, Subunit/immunology
- Vaccines, Subunit/genetics
- Vaccines, Subunit/administration & dosage
- Cytomegalovirus/immunology
- Cytomegalovirus/genetics
- Mice
- Viral Matrix Proteins/immunology
- Viral Matrix Proteins/genetics
- Cytomegalovirus Infections/prevention & control
- Cytomegalovirus Infections/immunology
- Humans
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Phosphoproteins/immunology
- Phosphoproteins/genetics
- Epitopes, T-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/genetics
- Epitopes, B-Lymphocyte/immunology
- Epitopes, B-Lymphocyte/genetics
- Female
- Immediate-Early Proteins/immunology
- Immediate-Early Proteins/genetics
- Antibodies, Viral/blood
- Mice, Inbred BALB C
- Immunogenicity, Vaccine
Collapse
Affiliation(s)
- Zonghui Li
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
- Department of Clinical Laboratory, Chengdu Aerotropolis Asia Heart Hospital, Chengdu, China
| | - Shasha Jiang
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
- Department of Clinical Laboratory, Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Wenxuan Liu
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Xiaoli Yang
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Fengjun Liu
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Xu Li
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Jun Li
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Meng Yu
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Zhun Wei
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, China
| | - Bin Wang
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Dongmeng Qian
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
19
|
Xie J, Wang H, Ma R, Fan J, Quan Q, Zhang Z, Li M, Li B. The molybdate transport protein ModA regulates nitrate reductase activity to increase the intestinal colonization and extraintestinal dissemination of Klebsiella pneumoniae in the inflamed gut. Virulence 2025; 16:2474185. [PMID: 40033924 PMCID: PMC11901421 DOI: 10.1080/21505594.2025.2474185] [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: 02/06/2024] [Revised: 01/24/2025] [Accepted: 02/25/2025] [Indexed: 03/05/2025] Open
Abstract
The mammalian intestine is a major site of colonization and a starting point of severe infections by Klebsiella pneumoniae. Inflammatory bowel disease (IBD) is an inflammatory disorder of the gut, and host-derived nitrate in IBD confers a luminal growth advantage upon Escherichia coli and Salmonella typhimurium through nitrate respiration in the inflamed gut. However, the impact of nitrate on the growth and pathogenicity of K. pneumoniae in this microenvironment is poorly understood. In this study, we used oral administration of dextran sodium sulphate to induce IBD in mouse models. We then analysed the colonization levels of K. pneumoniae wild-type (WT), the nitrate reductase gene mutant strains (ΔnarG, ΔnarZ and ΔnarGΔnarZ), and the molybdate uptake gene mutant strain (ΔmodA) in the inflamed intestinal tract. Results showed that the growth, intestinal colonization, and extraintestinal dissemination of K. pneumoniae were increased in the intestines of dextran sulphate sodium (DSS)-treated mice. Nitrate in the inflamed bowel conferred a growth advantage to K. pneumoniae through nitrate respiration. The molybdate transport protein ModA regulated nitrate reductase activity to increase the growth, intestinal colonization, and extraintestinal dissemination of K. pneumoniae. Tungstate will be a promising antibacterial agent to tackle K. pneumoniae infections in IBD patients.
Collapse
Affiliation(s)
- Jichen Xie
- School of Basic Medical Science, Hubei University of Medicine, Shiyan, China
| | - Hui Wang
- School of Basic Medical Science, Hubei University of Medicine, Shiyan, China
| | - Renhui Ma
- School of Basic Medical Science, Hubei University of Medicine, Shiyan, China
| | - Jinming Fan
- School of Basic Medical Science, Hubei University of Medicine, Shiyan, China
| | - Qiuhang Quan
- School of Basic Medical Science, Hubei University of Medicine, Shiyan, China
| | - Zhiqiang Zhang
- School of Basic Medical Science, Hubei University of Medicine, Shiyan, China
| | - Moran Li
- School of Basic Medical Science, Hubei University of Medicine, Shiyan, China
- Department of Respiratory, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Bei Li
- School of Basic Medical Science, Hubei University of Medicine, Shiyan, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, China
- Department of obstetricsl, Maternal and Child Health Hospital, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
20
|
Wang Y, Chen Y, Xiao Z, Shi Y, Fu C, Cao Y. Fecal microbiota transplantation modulates myeloid-derived suppressor cells and attenuates renal fibrosis in a murine model. Ren Fail 2025; 47:2480749. [PMID: 40141007 PMCID: PMC11951334 DOI: 10.1080/0886022x.2025.2480749] [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] [Revised: 03/07/2025] [Accepted: 03/09/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Renal fibrosis is a hallmark of progressive chronic kidney disease (CKD), with emerging evidence linking gut microbiota dysbiosis to disease progression. Myeloid-derived suppressor cells (MDSCs) have demonstrated renoprotective effects, yet the impact of fecal microbiota transplantation (FMT) on MDSC-mediated modulation of renal fibrosis remains unclear. METHODS C57BL/6J mice underwent unilateral ureteral obstruction (UUO) to induce renal fibrosis, followed by FMT administration via gavage. Flow cytometry was used to quantify granulocytic (G-MDSCs) and monocytic (M-MDSCs) MDSC populations in peripheral blood, kidney, and spleen. To elucidate the role of MDSCs in FMT-mediated effects, MDSCs were depleted or adoptively transferred in vivo. Renal fibrosis severity and inflammatory cytokine expression were subsequently analyzed. RESULTS FMT altered MDSC distribution, increasing M-MDSC accumulation in the blood and kidney. This was associated with downregulation of proinflammatory cytokines and attenuation of renal fibrosis. Adoptive MDSC transfer similarly produced anti-inflammatory and antifibrotic effects, reinforcing their therapeutic role in FMT-mediated renal protection. CONCLUSIONS FMT enhances M-MDSC-mediated immunomodulation, reducing inflammation and renal fibrosis in UUO-induced CKD. These findings suggest a potential therapeutic strategy targeting the gut-kidney axis in CKD management.
Collapse
Affiliation(s)
- Yajie Wang
- Department of Nephrology, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, China
- Anesthesia Laboratory and Training Center of Wannan Medical College, Wuhu, Anhui, China
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution (Wannan Medical College), Wuhu, Anhui, China
| | - Yuye Chen
- Department of Nephrology, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Zihao Xiao
- Department of Nephrology, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Yuanhui Shi
- Department of Nephrology, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Cong Fu
- Department of Cardiology, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Yuhan Cao
- Department of Nephrology, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, China
| |
Collapse
|
21
|
Woo S, Park PG, An T, Fatima M, Moon YE, Lee SY, Youn H, Hong KJ. Mini-review on the therapeutic vaccines targeting chronic infectious diseases: Evaluation system of therapeutic vaccines targeting HPV and EBV-related cancers. Hum Vaccin Immunother 2025; 21:2457187. [PMID: 39957237 PMCID: PMC11834422 DOI: 10.1080/21645515.2025.2457187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/05/2025] [Accepted: 01/17/2025] [Indexed: 02/18/2025] Open
Abstract
Chronic infectious diseases are threatening human health today, and their public health severity is increasing. The efficacy issues of drugs and the increase in drug-resistant pathogens require new response strategies for chronic infectious diseases, and therapeutic vaccines have recently been proposed as an effective alternative. However, research on therapeutic vaccines is still relatively underdeveloped. To solve this problem, an accurate understanding of the status and the challenge at hand of therapeutic vaccines targeting chronic infectious diseases is needed. In the present review, we provide an overview of the latest research trends in therapeutic vaccines targeting chronic infectious diseases and summarize the development status of therapeutic vaccines currently undergoing clinical research, focusing on the cases of human papillomavirus (HPV) and Epstein-Barr virus (EBV) as representative examples. We highlight the importance of standard methods for the evaluation of therapeutic vaccine, focusing on the cell-mediated immune response, which might accelerate therapeutic vaccine development.
Collapse
Affiliation(s)
- Seungkyun Woo
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Korea
| | - Pil-Gu Park
- Department of Microbiology, Gachon University College of Medicine, Incheon, Korea
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea
- Korea mRNA Vaccine Initiative, Gachon University, Seongnam, Korea
| | - Timothy An
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea
| | - Munazza Fatima
- Department of Microbiology, Gachon University College of Medicine, Incheon, Korea
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea
| | - Ye-Eun Moon
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Korea
| | - Seok-Yong Lee
- Department of Nuclear Medicine, Cancer Imaging Center, Seoul National University Hospital, Seoul, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hyewon Youn
- Department of Nuclear Medicine, Cancer Imaging Center, Seoul National University Hospital, Seoul, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Kee-Jong Hong
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Korea
- Department of Microbiology, Gachon University College of Medicine, Incheon, Korea
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea
- Korea mRNA Vaccine Initiative, Gachon University, Seongnam, Korea
| |
Collapse
|
22
|
Emami S, Westerlund E, Rojas Converso T, Johansson-Lindbom B, Persson JJ. Protection acquired upon intraperitoneal group a Streptococcus immunization is independent of concurrent adaptive immune responses but relies on macrophages and IFN-γ. Virulence 2025; 16:2457957. [PMID: 39921669 PMCID: PMC11810095 DOI: 10.1080/21505594.2025.2457957] [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: 08/09/2024] [Revised: 11/09/2024] [Accepted: 01/20/2025] [Indexed: 02/10/2025] Open
Abstract
Group A Streptococcus (GAS; Streptococcus pyogenes) is an important bacterial pathogen causing over 700 million superficial infections and around 500.000 deaths due to invasive disease or severe post-infection sequelae yearly. In spite of this major impact on society, there is currently no vaccine available against this bacterium. GAS strains can be separated into >250 distinct emm (M)-types, and protective immunity against GAS is believed to in part be dependent on type-specific antibodies. Here, we analyse the nature of protective immunity generated against GAS in a model of intraperitoneal immunization in mice. We demonstrate that multiple immunizations are required for the ability to survive a subsequent lethal challenge, and although significant levels of GAS-specific antibodies are produced, these are redundant for protection. Instead, our data show that the immunization-dependent protection in this model is induced in the absence of B and T cells and is accompanied by the induction of an altered acute cytokine profile upon subsequent infection, noticeable e.g. by the absence of classical pro-inflammatory cytokines and increased IFN-γ production. Further, the ability of immunized mice to survive a lethal infection is dependent on macrophages and the macrophage-activating cytokine IFN-γ. To our knowledge these findings are the first to suggest that GAS may have the ability to induce forms of trained innate immunity. Taken together, the current study proposes a novel role for the innate immune system in response to GAS infections that potentially could be leveraged for future development of effective vaccines.
Collapse
Affiliation(s)
- Shiva Emami
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Elsa Westerlund
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | | | - Jenny J Persson
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
23
|
Moutsoglou D, Ramakrishnan P, Vaughn BP. Microbiota transplant therapy in inflammatory bowel disease: advances and mechanistic insights. Gut Microbes 2025; 17:2477255. [PMID: 40062406 PMCID: PMC11901402 DOI: 10.1080/19490976.2025.2477255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/27/2025] [Accepted: 03/04/2025] [Indexed: 03/14/2025] Open
Abstract
Microbiota transplant therapy is an emerging therapy for inflammatory bowel disease, but factors influencing its efficacy and mechanism remain poorly understood. In this narrative review, we outline key elements affecting therapeutic outcomes, including donor factors (such as age and patient relationship), recipient factors, control selection, and elements impacting engraftment and its correlation with clinical response. We also examine potential mechanisms through inflammatory bowel disease trials, focusing on the interplay between the microbiota, host, and immune system. Finally, we briefly explore potential future directions for microbiota transplant therapy and promising emerging treatments.
Collapse
Affiliation(s)
- Daphne Moutsoglou
- Gastroenterology Section, Minneapolis VA Health Care System, Minneapolis, MN, USA
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | | | - Byron P. Vaughn
- Division of Gastroenterology, Hepatology, and Nutrition, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
24
|
Swain J, Askenasy I, Rudland Nazeer R, Ho PM, Labrini E, Mancini L, Xu Q, Hollendung F, Sheldon I, Dickson C, Welch A, Agbamu A, Godlee C, Welch M. Pathogenicity and virulence of Pseudomonas aeruginosa: Recent advances and under-investigated topics. Virulence 2025; 16:2503430. [PMID: 40353451 PMCID: PMC12087490 DOI: 10.1080/21505594.2025.2503430] [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: 01/31/2025] [Revised: 04/23/2025] [Accepted: 05/03/2025] [Indexed: 05/14/2025] Open
Abstract
Pseudomonas aeruginosa is a model for the study of quorum sensing, protein secretion, and biofilm formation. Consequently, it has become one of the most intensely reviewed pathogens, with many excellent articles in the current literature focusing on these aspects of the organism's biology. Here, though, we aim to take a slightly different approach and consider some less well appreciated (but nonetheless important) factors that affect P. aeruginosa virulence. We start by reminding the reader of the global importance of P. aeruginosa infection and that the "virulome" is very niche-specific. Overlooked but obvious questions such as "what prevents secreted protein products from being digested by co-secreted proteases?" are discussed, and we suggest how the nutritional preference(s) of the organism might dictate its environmental reservoirs. Recent studies identifying host genes associated with genetic predisposition towards P. aeruginosa infection (and even infection by specific P. aeruginosa strains) and the role(s) of intracellular P. aeruginosa are introduced. We also discuss the fact that virulence is a high-risk strategy and touch on how expression of the two main classes of virulence factors is regulated. A particular focus is on recent findings highlighting how nutritional status and metabolism are as important as quorum sensing in terms of their impact on virulence, and how co-habiting microbial species at the infection site impact on P. aeruginosa virulence (and vice versa). It is our view that investigation of these issues is likely to dominate many aspects of research into this WHO-designated priority pathogen over the next decade.
Collapse
Affiliation(s)
- Jemima Swain
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | - Isabel Askenasy
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | | | - Pok-Man Ho
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | - Edoardo Labrini
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | | | - Qingqing Xu
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | | | | | - Camilla Dickson
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | - Amelie Welch
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | - Adam Agbamu
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | - Camilla Godlee
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | - Martin Welch
- Department of Biochemistry, Cambridge University, Cambridge, UK
| |
Collapse
|
25
|
Gupta A, Maffulli N. Undenatured type II collagen for knee osteoarthritis. Ann Med 2025; 57:2493306. [PMID: 40253594 PMCID: PMC12010644 DOI: 10.1080/07853890.2025.2493306] [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: 01/10/2024] [Revised: 08/23/2024] [Accepted: 04/09/2025] [Indexed: 04/22/2025] Open
Abstract
INTRODUCTION Knee Osteoarthritis (OA) leads to significant pain and reduced function and affects patients' overall quality of life (QoL). Conservative modalities are the first line of management, resorting to surgery only if they fail. However, these modalities have limitations, and do not address the underlying cause of knee OA. The use of nutraceuticals, including native/undenatured type II collagen (UC-2), has evolved and shown promise in the conservative management of knee OA. This article highlights the mechanism of action, and qualitatively presents the pre-clinical, clinical and on-going scientific literature exploring the safety and efficacy of UC-2 for the management of knee OA. METHODS A search was performed using multiple databases (PubMed, Web of Science, Embase and Scopus) employing terms for UC-2 and Knee OA for articles published in English language, while adhering to Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines. All pre-clinical and clinical studies utilizing UC-2 for knee OA were included. Studies not using UC-2 alone or not focusing on the management of knee OA were excluded. RESULTS Twelve studies (3 pre-clinical studies, 8 clinical studies and 1 study with both pre-clinical and clinical component) met our pre-defined search and inclusion criteria, and were included in this review. DISCUSSION UC-2 acts via a specific immune mediated mechanism, known as oral tolerance, which can lead to reduced inflammation and enhanced cartilage repair in the knee joint. In addition, administration of UC-2 (40 mg daily) is safe and efficacious in the short- and mid-term, reducing inflammation and pain, and improving function, range of motion (ROM) and overall QoL. Nonetheless, more adequately powered, prospective, multi-center, non-randomized and randomized controlled trials with longer follow-up are warranted to establish the long-term efficacy of UC-2 in knee OA patients and justify its routine clinical use.
Collapse
Affiliation(s)
| | - Nicola Maffulli
- Department of Musculoskeletal Disorders, School of Medicine and Surgery, University of Salerno, Fisciano, Italy
- Department of Trauma and Orthopaedics, Ospedale Sant’ Andrea, Sapienza University of Rome, Rome, Italy
- Barts and the London School of Medicine and Dentistry, Centre for Sports and Exercise Medicine, Queen Mary University of London, London, UK
- School of Pharmacy and Bioengineering, Keele University School of Medicine, Stoke on Trent, UK
| |
Collapse
|
26
|
Sun X. Dilemma in prevention of pertussis infection among infants under six months in China. Expert Rev Vaccines 2025; 24:138-145. [PMID: 39869378 DOI: 10.1080/14760584.2025.2459745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 01/19/2025] [Accepted: 01/24/2025] [Indexed: 01/28/2025]
Abstract
INTRODUCTION Pertussis poses a significant threat to infants under six months due to their immature immune systems, limited maternal antibody protection, and constraints in the vaccination schedule. Despite vaccination efforts, this group remains highly susceptible to severe complications. Addressing these challenges is crucial for improving the health outcomes of infants in China. AREAS COVERED This review examines the primary challenges in preventing pertussis infections among infants under six months in China, focusing on factors such as underdeveloped immune system and inadequate maternal antibody protection. It analyzes limitations in current vaccination strategies and the impact of socio-cultural factors, healthcare resource distribution, and surveillance inadequacies. A comprehensive literature search was conducted to identify potential solutions, including enhancing maternal immunization, adjusting early vaccination strategies, increasing vaccine coverage, and developing new vaccines. The review synthesizes current research findings and data to provide a detailed overview of these issues. EXPERT OPINION Infants under six months are particularly vulnerable to pertussis. Early and effective prevention strategies, such as enhanced maternal immunization and adjusted vaccination schedules, are needed. Increasing vaccine coverage and developing safer, more immunogenic vaccines are essential. Policymakers should prioritize these measures to reduce pertussis incidence and complications among infants in China.
Collapse
Affiliation(s)
- Xiang Sun
- Department of Expanded Program on Immunization, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| |
Collapse
|
27
|
Mura M, Trignol A, Le Dault E, Tournier JN. Lessons for medical countermeasure development from unforeseen outbreaks. Emerg Microbes Infect 2025; 14:2471035. [PMID: 39976365 PMCID: PMC11894751 DOI: 10.1080/22221751.2025.2471035] [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: 11/06/2024] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 02/21/2025]
Abstract
The unanticipated emergence of the COVID-19 pandemic and the rapid spread of the mpox epidemic in 2022 and 2024 brought unforeseen challenges to public health. While distinct in nature, these outbreaks share some similarities and offer valuable insights into responding to novel virus dissemination in vulnerable populations. In light of these two experiences, we aim to discern the prioritization of medical countermeasures (MCM) among antivirals, antibodies, and vaccines. Comparative analysis of MCMs reveals that while antivirals serve essential roles as therapeutic tools, monoclonal antibodies can be used for both prevention and treatment, and vaccines remain of paramount importance for controlling epidemics as mass or targeted prophylaxis. Variability in production processes, administration methods, logistics, and costs distinguish these countermeasures. Vaccines, by inducing long-lasting immunity and ideally promoting herd effects, exhibit substantial advantages over other options. To enhance future pandemic readiness, proactive measures must include ready-to-use vaccine platforms with regulatory approval and manufacturing capacities, as well as prototype vaccines for representative pathogens and preexisting protocols to evaluate their efficacies and side effects. The comparison underscores the challenges of social acceptance and equity, particularly in vaccine production and distribution. As the world faces unknown agents, the three major types of MCMs do not have equal and symmetrical effects in terms of epidemic control. Thus, a vaccine-oriented strategy with a community-centered approach, proves essential for effective pandemic preparedness, encouraging continued innovation in vaccinology.
Collapse
Affiliation(s)
- Marie Mura
- Microbiology and Infectious Diseases Department, Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge Cedex, France
| | - Aurélie Trignol
- Microbiology and Infectious Diseases Department, Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge Cedex, France
| | - Erwan Le Dault
- Microbiology and Infectious Diseases Department, Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge Cedex, France
- Department of Infectious Diseases and Tropical Medicine, Laveran Military Teaching Hospital, Marseille, France
| | - Jean-Nicolas Tournier
- Microbiology and Infectious Diseases Department, Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge Cedex, France
- École du Val-de-Grâce, Paris, France
| |
Collapse
|
28
|
Chen Y, Li Y, Xu Y, Lv Q, Ye Y, Gu J. Revealing the role of natural killer cells in ankylosing spondylitis: identifying diagnostic biomarkers and therapeutic targets. Ann Med 2025; 57:2457523. [PMID: 39853176 PMCID: PMC11770870 DOI: 10.1080/07853890.2025.2457523] [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: 01/22/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND Ankylosing spondylitis (AS) is a chronic autoimmune disease that primarily affects the axial joints. Immune cells play a key role in the pathogenesis of AS. This study integrated bioinformatics methods with experimental validation to explore the role of natural killer (NK) cells in AS. METHODS Two microarray datasets, GSE25101 and GSE73754, were selected, and the scRNA-seq data were obtained from GSE194315 and Liu's research. Differentially expressed genes (DEGs) and functional enrichment analysis were performed respectively. Weighted gene co-expression network analysis (WGCNA) was conducted to identify key modules of co-expressed genes and genes involved in NK cell function. The diagnostic value of the identified key genes was evaluated using ROC curves, logistic regression analysis, and a nomogram. Real-time PCR (RT-PCR) was used to quantified the expression of genes. Statistical analysis was conducted using the R software package, and a p-value of less than 0.05 was considered statistically significant. RESULTS Pathways enrichment analysis revealed the involvement of NK cell-mediated immune pathways and regulation of the innate immune response, indicating the crucial role of innate immunity, especially NK cells, in AS pathogenesis. The construction of a co-expression network revealed that the MElightyellow module was most relevant to the NK cell-mediated immune pathway. IL2RB, CD247, PLEKHF1, EOMES, S1PR5, FGFBP2 from the MElightyellow module were identified as key genes involved in NK cell-mediated immune response and served as potential diagnostic biomarkers for AS, with moderate to high diagnostic values based on AUC values. Further analysis using scRNA-seq profiling revealed the higher expression level of IL2RB, CD247, PLEKHF1, S1PR5, FGFBP2 in NK cells compared to that in other cell types. CD247, PLEKHF1, EOMES, S1PR5, and FGFBP2 were reduced expressed in AS patients as compare to control group verified by scRNA-seq data, CD247, EOMES, FGFBP2, IL2RB and S1PR5 were reduced expressed verified by RT-PCR, and PLEKHF1, S1PR5, and FGFBP2 was upregulated after TNF-α blocker therapy. CONCLUSION The study revealed the potential role of NK cells and identified IL2RB, CD247, PLEKHF1, EOMES, S1PR5, and FGFBP2 as key genes associated with NK cells in the pathogenesis of AS.
Collapse
Affiliation(s)
- Yuling Chen
- Department of Rheumatology and Immunology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong Province, People’s Republic of China
| | - Yan Li
- Department of Scientific Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong Province, People’s Republic of China
| | - Yuan Xu
- Department of Clinical Laboratory, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong Province, People’s Republic of China
| | - Qing Lv
- Department of Rheumatology and Immunology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong Province, People’s Republic of China
| | - Yuanchun Ye
- School of Science, Shenzhen Campus of Sun Yat-sen University, Shenzhen, People’s Republic of China
| | - Jieruo Gu
- Department of Rheumatology and Immunology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong Province, People’s Republic of China
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong ProvincePeople’s Republic of China
| |
Collapse
|
29
|
Sweet-Jones J, Martin AC. An antibody developability triaging pipeline exploiting protein language models. MAbs 2025; 17:2472009. [PMID: 40038849 PMCID: PMC11901365 DOI: 10.1080/19420862.2025.2472009] [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: 10/31/2024] [Revised: 02/17/2025] [Accepted: 02/20/2025] [Indexed: 03/06/2025] Open
Abstract
Therapeutic monoclonal antibodies (mAbs) are a successful class of biologic drugs that are frequently selected from phage display libraries and transgenic mice that produce fully human antibodies. However, binding affinity to the correct epitope is necessary, but not sufficient, for a mAb to have therapeutic potential. Sequence and structural features affect the developability of an antibody, which influences its ability to be produced at scale and enter trials, or can cause late-stage failures. Using data on paired human antibody sequences, we introduce a pipeline using a machine learning approach that exploits protein language models to identify antibodies which cluster with antibodies that have entered the clinic and are therefore expected to have developability features similar to clinically acceptable antibodies, and triage out those without these features. We propose this pipeline as a useful tool in candidate selection from large libraries, reducing the cost of exploration of the antibody space, and pursuing new therapeutics.
Collapse
Affiliation(s)
- James Sweet-Jones
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London, UK
| | - Andrew C.R. Martin
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London, UK
| |
Collapse
|
30
|
Kamble NS, Thomas S, Madaan T, Ehsani N, Sange S, Tucker K, Muhumure A, Kunkler S, Kotagiri N. Engineered bacteria as an orally administered anti-viral treatment and immunization system. Gut Microbes 2025; 17:2500056. [PMID: 40340796 PMCID: PMC12064065 DOI: 10.1080/19490976.2025.2500056] [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: 01/29/2025] [Revised: 04/05/2025] [Accepted: 04/24/2025] [Indexed: 05/10/2025] Open
Abstract
The emergence of new viral pathogens necessitates innovative antiviral therapies and vaccines. Traditional approaches, such as monoclonal antibodies and vaccines, are often hindered by resistance, limited effectiveness, and high costs. Here, we develop an engineered probiotic-based antiviral platform using Escherichia coli Nissle 1917 (EcN), capable of providing both mucosal and systemic immunity via oral administration. EcN was engineered to display anti-spike nanobodies or express the Spike-Receptor Binding Domain on its surface. Our findings reveal that EcN with nanobodies effectively inhibits the interaction between spike protein-expressing pseudoviruses and the ACE2 receptor. Furthermore, we observed the translocation of nanobodies to distant organs, facilitated by outer membrane vesicles (OMVs). The oral administration of EcN expressing spike proteins induced a robust immune response characterized by the production of both IgG and IgA, antibodies that blocked the pseudovirus-ACE2 interaction. While SARS-CoV-2 served as a model, this versatile probiotic platform holds potential for developing customizable biotherapeutics against a wide range of emerging pathogens such as influenza virus or respiratory syncytial virus (RSV) by engineering EcN to express viral surface protein or neutralizing nanobodies demonstrating its versatility as a next-generation mucosal vaccine strategy.
Collapse
Affiliation(s)
- Nitin S. Kamble
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | - Shindu Thomas
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | - Tushar Madaan
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | - Nadia Ehsani
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | - Saqib Sange
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | - Kiersten Tucker
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | - Alexis Muhumure
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | - Sarah Kunkler
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | - Nalinikanth Kotagiri
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
31
|
Warang P, Singh G, Moshir M, Binazon O, Laghlali G, Chang LA, Wouters H, Vanhoenacker P, Notebaert M, Elhemdaoui N, Augustynková K, Steeland S, Ulrichts P, Baumeister J, Schotsaert M. Impact of FcRn antagonism on vaccine-induced protective immune responses against viral challenge in COVID-19 and influenza mouse vaccination models. Hum Vaccin Immunother 2025; 21:2470542. [PMID: 40028815 PMCID: PMC11881870 DOI: 10.1080/21645515.2025.2470542] [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: 08/20/2024] [Revised: 02/03/2025] [Accepted: 02/19/2025] [Indexed: 03/05/2025] Open
Abstract
Antagonism of the neonatal Fc receptor through an engineered antibody Fc fragment, such as efgartigimod, results in a decrease in immunoglobulin G levels. This approach is being evaluated as a therapeutic strategy for the treatment of IgG-mediated autoimmune diseases. Our goal was to evaluate the impact of mFc-ABDEG, a mouse-adapted antibody Fc fragment with a mode of action highly similar to efgartigimod, on vaccine-induced protective immune responses against viral infections. Therefore, mouse vaccination models for COVID-19 and influenza were employed, utilizing an mRNA COVID-19 vaccine (COMIRNATY) and an adjuvanted, inactivated quadrivalent influenza vaccine (Seqirus+AddaVax), respectively. In both models, vaccination induced robust humoral responses. As expected, animals treated with mFc-ABDEG had lower levels of virus-specific IgG, while virus-specific IgM responses remained unaffected. The COVID-19 vaccine induced a strong Th1-type T cell response irrespective of mFc-ABDEG treatment. Influenza vaccination resulted in a poor T cell induction, regardless of mFc-ABDEG treatment, due to the Th2-biased response that inactivated influenza vaccines typically induce. Importantly, mFc-ABDEG treatment had no effect on protective immunity against live viral challenges in both models. Vaccinated animals treated with mFc-ABDEG were equally protected as the non-treated vaccinated controls. These non-clinical data demonstrate that FcRn antagonism with mFc-ABDEG did not affect the generation of vaccine-induced protective humoral and cellular responses, or protection against viral challenges. These data substantiate the clinical observations that, although IgG titers were reduced, FcRn antagonism with efgartigimod did not impair the ability to generate new specific IgG responses, regardless of the timing of vaccination.
Collapse
Affiliation(s)
- Prajakta Warang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gagandeep Singh
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mahan Moshir
- Department of Translational & Clinical Sciences, Argenx, Ghent, Belgium
| | - Ornella Binazon
- Department of Non-Clinical Pharmacology & Toxicology, Argenx, Ghent, Belgium
| | - Gabriel Laghlali
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lauren A. Chang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | | | | | | | - Sophie Steeland
- Department of Translational & Clinical Sciences, Argenx, Ghent, Belgium
| | - Peter Ulrichts
- Department of Translational & Clinical Sciences, Argenx, Ghent, Belgium
| | - Judith Baumeister
- Department of Non-Clinical Pharmacology & Toxicology, Argenx, Ghent, Belgium
| | - Michael Schotsaert
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
32
|
Lv Y, Yang X, Sun X, Ren X. Immune-microbiota dysregulation in maintenance hemodialysis: a 16S rRNA sequencing-based analysis of gut flora and T cell profiles. Ren Fail 2025; 47:2498630. [PMID: 40375064 PMCID: PMC12082729 DOI: 10.1080/0886022x.2025.2498630] [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: 01/24/2025] [Revised: 04/13/2025] [Accepted: 04/19/2025] [Indexed: 05/18/2025] Open
Abstract
BACKGROUND Maintenance hemodialysis (MHD) patients frequently exhibit immune dysregulation and gut dysbiosis, both of which contribute to increased infection risk and adverse outcomes. However, the relationship between gut microbial composition and immune competence in this population remains underexplored. METHODS This study assessed 45 MHD patients and 30 healthy controls, stratifying MHD patients into immunocompetent (HD-NLI, CD4+/CD8+ ≥ 1) and immunodeficient (HD-LI, CD4+/CD8+ < 1) groups. Circulating cytokines (IL-6, IL-10, IL-12, TNF-α, IFN-γ) were quantified using ELISA. Gut microbiota profiles were derived via 16S rRNA gene sequencing (V3-V4 regions), followed by QIIME2 and LEfSe-based bioinformatics analyses. RESULTS HD-LI patients displayed severe T cell dysregulation and elevated pro-inflammatory cytokines. Compared to controls, HD patients had reduced abundance of beneficial taxa (e.g., Prevotella copri, Bacteroides vulgatus, Agathobacter), and enrichment of pro-inflammatory taxa (e.g., Escherichia-Shigella, Blautia, Citrobacter). LEfSe identified 39 discriminatory taxa with distinct immune group signatures. Redundancy analysis revealed that CD4+ levels, CD4+/CD8+ ratios, and TNF-α significantly shaped microbiota composition. Correlation analysis confirmed strong associations between immune parameters and microbial taxa involved in short-chain fatty acid (SCFA) metabolism. CONCLUSION This study provides novel evidence linking gut microbial dysbiosis to immune impairment in MHD patients. The findings suggest that SCFA-producing bacteria are depleted in immunodeficient states, offering a potential target for microbiota-directed immunomodulatory therapies in ESRD.
Collapse
Affiliation(s)
- Yan Lv
- Department of Nephrology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiuting Yang
- Department of Epidemiology, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Xiaowu Sun
- Department of Nephrology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohong Ren
- Department of Intensive Care Unit, Lvliang People’s Hospital, Lvliang City, China
| |
Collapse
|
33
|
Xu Z, Qiao S, Wang Z, Peng C, Hou Y, Liu B, Cao G, Wang T. PMA1-containing extracellular vesicles of Candida albicans triggers immune responses and colitis progression. Gut Microbes 2025; 17:2455508. [PMID: 39886799 PMCID: PMC11792855 DOI: 10.1080/19490976.2025.2455508] [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: 10/14/2024] [Revised: 01/08/2025] [Accepted: 01/13/2025] [Indexed: 02/01/2025] Open
Abstract
Candida albicans (C. albicans) exhibits aberrant changes in patients with colitis, and it has been reported to dominate the colonic mucosal immune response. Here, we found that PMA1 expression was significantly increased in C. albicans from patients with IBD compared to that in healthy controls. A Crispr-Cas9-based fungal strain editing system was then used to knock out PMA1 expression in C. albicans. Compared to WT-C.a, ΔPMA1-C.a could not aggravate colitis. Proteomic analysis showed that PMA1 was transported by extracellular vesicles (EVs) of C. albicans. PMA1-containing EVs aggravated colitis, modulated the migration of cDC2 from the lamina propria to mesenteric lymph nodes, and induced TH17 cell differentiation. Moreover, the adaptor protein CARD9 was critical in PMA1-containing EV-induced colitis, and CARD9-deficient DCs did not induce TH17 cell differentiation or IL-17A production. Mechanically, CARD9 combines with the glycolytic protein GAPDH (aa2-146 domain) through its CARD region. CARD9 deficiency led to decreased enzyme activity of GAPDH and decreased glycolysis of DCs. These findings indicate that PMA1 is a potential virulence factor responsible for the pathogenesis of C. albicans colitis.
Collapse
Affiliation(s)
- Zhen Xu
- Department of Oncology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing, China
| | - Shuping Qiao
- Department of Oncology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing, China
| | - Zelin Wang
- Department of Oncology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing, China
| | - Chen Peng
- Department of Oncology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing, China
| | - Yayi Hou
- Department of Oncology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing, China
| | - Baorui Liu
- Department of Oncology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Guochun Cao
- Department of Medical Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Tingting Wang
- Department of Oncology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
34
|
Lin A, Jiang A, Huang L, Li Y, Zhang C, Zhu L, Mou W, Liu Z, Zhang J, Cheng Q, Wei T, Luo P. From chaos to order: optimizing fecal microbiota transplantation for enhanced immune checkpoint inhibitors efficacy. Gut Microbes 2025; 17:2452277. [PMID: 39826104 DOI: 10.1080/19490976.2025.2452277] [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: 10/16/2024] [Revised: 11/22/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025] Open
Abstract
The integration of fecal microbiota transplantation (FMT) with immune checkpoint inhibitors (ICIs) presents a promising approach for enhancing cancer treatment efficacy and overcoming therapeutic resistance. This review critically examines the controversial effects of FMT on ICIs outcomes and elucidates the underlying mechanisms. We investigate how FMT modulates gut microbiota composition, microbial metabolite profiles, and the tumor microenvironment, thereby influencing ICIs effectiveness. Key factors influencing FMT efficacy, including donor selection criteria, recipient characteristics, and administration protocols, are comprehensively discussed. The review delineates strategies for optimizing FMT formulations and systematically monitoring post-transplant microbiome dynamics. Through a comprehensive synthesis of evidence from clinical trials and preclinical studies, we elucidate the potential benefits and challenges of combining FMT with ICIs across diverse cancer types. While some studies report improved outcomes, others indicate no benefit or potential adverse effects, emphasizing the complexity of host-microbiome interactions in cancer immunotherapy. We outline critical research directions, encompassing the need for large-scale, multi-center randomized controlled trials, in-depth microbial ecology studies, and the integration of multi-omics approaches with artificial intelligence. Regulatory and ethical challenges are critically addressed, underscoring the imperative for standardized protocols and rigorous long-term safety assessments. This comprehensive review seeks to guide future research endeavors and clinical applications of FMT-ICIs combination therapy, with the potential to improve cancer patient outcomes while ensuring both safety and efficacy. As this rapidly evolving field advances, maintaining a judicious balance between openness to innovation and cautious scrutiny is crucial for realizing the full potential of microbiome modulation in cancer immunotherapy.
Collapse
Affiliation(s)
- Anqi Lin
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong, China
| | - Aimin Jiang
- Department of Urology, Changhai hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Lihaoyun Huang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong, China
| | - Yu Li
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong, China
| | - Chunyanx Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong, China
| | - Lingxuan Zhu
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong, China
| | - Weiming Mou
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong, China
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zaoqu Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Hunan, China
| | - Ting Wei
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong, China
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, 999078, China
| |
Collapse
|
35
|
Zöggeler T, Kavallar AM, Pollio AR, Aldrian D, Decristoforo C, Scholl-Bürgi S, Müller T, Vogel GF. Meta-analysis of shotgun sequencing of gut microbiota in obese children with MASLD or MASH. Gut Microbes 2025; 17:2508951. [PMID: 40396204 DOI: 10.1080/19490976.2025.2508951] [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: 07/31/2024] [Revised: 03/25/2025] [Accepted: 05/14/2025] [Indexed: 05/22/2025] Open
Abstract
Alterations in the gut microbiome affect the development and severity of metabolic dysfunction-associated steatotic liver disease (MASLD) or metabolic dysfunction-associated steatohepatitis (MASH). We analyzed microbiomes of obese children with and without MASLD, MASH, and healthy controls. Electronic databases were searched for studies on the gut microbiome in children with obesity with/without MASLD or MASH, providing shotgun-metagenomic-sequencing data. Nine studies and an additionally recruited cohort were included. Fecal microbiomes of children with MASLD (n = 153) and MASH (n = 70) were significantly different in alpha- and beta-diversity (p < 0.001) compared to obese (n = 58) and healthy (n = 132). Species Faecalibacterium_prausnitzii and Prevotella_copri are differentially abundant between obese, MASLD and MASH groups. XGBoost and random forest-models accurately predict MASLD over obesity with an AUROC of 87% and MASH over MASLD with 89%. Pathway-abundance-based models accurately predict MASLD over obesity with an AUROC of 81% and MASH over MASLD with 88%. The composition of the gut microbiome is altered with increasing hepatic fibrosis and concomitant species-abundance increase of Prevotella_copri (p = 0.0082). Machine-learning models discriminate pediatric from adult MASH with an AUROC of 97%. The gut microbial composition is increasingly altered in children with the progression of MASLD toward MASH. This can be utilized as a fecal biomarker and highlights the impact of diet on the gut microbiome for disease intervention.
Collapse
Affiliation(s)
- Thomas Zöggeler
- Department of Paediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Anna Maria Kavallar
- Department of Paediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Adam Robert Pollio
- Institute of Cell Biology, Medical University of Innsbruck, Innsbruck, Austria
| | - Denise Aldrian
- Department of Paediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Sabine Scholl-Bürgi
- Department of Paediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Thomas Müller
- Department of Paediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Georg Friedrich Vogel
- Department of Paediatrics I, Medical University of Innsbruck, Innsbruck, Austria
- Institute of Cell Biology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
36
|
Zheng X, Liu B, Ni P, Cai L, Shi X, Ke Z, Zhang S, Hu B, Yang B, Xu Y, Long W, Fang Z, Wang Y, Zhang W, Xu Y, Wang Z, Pan K, Zhou K, Wang H, Geng H, Hu H, Liu B. Development and application of an uncapped mRNA platform. Ann Med 2025; 57:2437046. [PMID: 39648715 PMCID: PMC11632943 DOI: 10.1080/07853890.2024.2437046] [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: 10/03/2023] [Revised: 11/28/2023] [Accepted: 06/01/2024] [Indexed: 12/10/2024] Open
Abstract
BACKGROUND A novel uncapped mRNA platform was developed. METHODS Five lipid nanoparticle (LNP)-encapsulated mRNA constructs were made to evaluate several aspects of our platform, including transfection efficiency and durability in vitro and in vivo and the activation of humoral and cellular immunity in several animal models. The constructs were eGFP-mRNA-LNP (for enhanced green fluorescence mRNA), Fluc-mRNA-LNP (for firefly luciferase mRNA), SδT-mRNA-LNP (for Delta strain SARS-CoV-2 spike protein trimer mRNA), gDED-mRNA-LNP (for truncated glycoprotein D mRNA coding ectodomain from herpes simplex virus type 2 (HSV2)) and gDFR-mRNA-LNP (for truncated HSV2 glycoprotein D mRNA coding amino acids 1-400). RESULTS Quantifiable target protein expression was achieved in vitro and in vivo with eGFP- and Fluc-mRNA-LNP. SδT-mRNA-LNP, gDED-mRNA-LNP and gDFR-mRNA-LNP induced both humoral and cellular immune responses comparable to those obtained by previously reported capped mRNA-LNP constructs. Notably, SδT-mRNA-LNP elicited neutralizing antibodies in hamsters against the Omicron and Delta strains. Additionally, gDED-mRNA-LNP and gDFR-mRNA-LNP induced potent neutralizing antibodies in rabbits and mice. The mRNA constructs with uridine triphosphate (UTP) outperformed those with N1-methylpseudouridine triphosphate (N1mψTP) in the induction of antibodies via SδT-mRNA-LNP. CONCLUSIONS Our uncapped, process-simplified and economical mRNA platform may have broad utility in vaccines and protein replacement drugs.KEY MESSAGESThe mRNA platform described in our paper uses internal ribosome entry site (IRES) (Rapid, Amplified, Capless and Economical, RACE; Register as BH-RACE platform) instead of caps and uridine triphosphate (UTP) instead of N1-methylpseudouridine triphosphate (N1mψTP) to synthesize mRNA.Through the self-developed packaging instrument and lipid nanoparticle (LNP) delivery system, mRNA can be expressed in cells more efficiently, quickly and economically.Particularly exciting is that potent neutralizing antibodies against Delta and Omicron real viruses were induced with the new coronavirus S protein mRNA vaccine from the BH-RACE platform.
Collapse
Affiliation(s)
- Xiaodi Zheng
- College of Bioengineering, National ‘‘111’’ Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei University of Technology, Wuhan, China
| | - Biao Liu
- College of Bioengineering, National ‘‘111’’ Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei University of Technology, Wuhan, China
| | - Peng Ni
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| | - Linkang Cai
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| | - Xiaotai Shi
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| | - Zonghuang Ke
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| | - Siqi Zhang
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| | - Bing Hu
- Hubei Provincial Centre for Disease Control and Prevention, Wuhan, China
| | - Binfeng Yang
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| | - Yiyan Xu
- College of Bioengineering, National ‘‘111’’ Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei University of Technology, Wuhan, China
| | - Wei Long
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| | - Zhizheng Fang
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| | - Yang Wang
- College of Bioengineering, National ‘‘111’’ Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei University of Technology, Wuhan, China
| | - Wen Zhang
- Department of Immunology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Xu
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| | - Zhong Wang
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| | - Kai Pan
- Hubei Provincial Centre for Disease Control and Prevention, Wuhan, China
| | - Kangping Zhou
- Hubei Provincial Centre for Disease Control and Prevention, Wuhan, China
| | - Hanming Wang
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| | - Hui Geng
- School of Life Science, Huazhong Normal University, Wuhan, China
| | - Han Hu
- College of Bioengineering, National ‘‘111’’ Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei University of Technology, Wuhan, China
| | - Binlei Liu
- College of Bioengineering, National ‘‘111’’ Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei University of Technology, Wuhan, China
- Wuhan Binhui Biopharmaceutical Co., Ltd., Wuhan, China
| |
Collapse
|
37
|
Haller R, Cai Y, de Buhr N, Rieder JC, Schlüter D, Baier C, Rohde H, von Köckritz-Blickwede M, Vital M, Winstel V. Transmissible Staphylococcus pseudintermedius thwarts neutrophil extracellular trap-driven containment to promote invasive disease. Emerg Microbes Infect 2025; 14:2482709. [PMID: 40172876 PMCID: PMC12001851 DOI: 10.1080/22221751.2025.2482709] [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: 12/13/2024] [Revised: 03/17/2025] [Accepted: 03/17/2025] [Indexed: 04/04/2025]
Abstract
Methicillin-resistant Staphylococcus pseudintermedius (MRSP) is an emerging zoonotic pathogen that causes a variety of clinical diseases in mammalian hosts. While it frequently causes infections in dogs and other domestic animals, accumulating evidence indicates that zoonotic spillover and cross-species transmission events favour local and invasive S. pseudintermedius infections in humans. However, immuno-evasive maneuvers that shape S. pseudintermedius pathogenicity and survival in diseased hosts remain enigmatic. Powered by multi-tech imaging and a mouse model of bloodstream infection, we illustrate that S. pseudintermedius adopted a virulence mechanism from predominant bacterial pathogens to surmount neutrophilic responses and neutrophil extracellular trap (NET)-mediated killing. Specifically, release of NucB, a thermostable nuclease, helps MRSP coping with the antimicrobial and pathogen-immobilizing properties of NETs and even promotes intra-neutrophil survival upon phagocytosis, thereby contributing to S. pseudintermedius pathogenesis and persistence within hepatic abscesses. Combined with the analysis of genetically distinct human clinical isolates, all of which display nuclease activity and features of resistance to NETosis-induced killing, our data highlight how zoonotic staphylococci overcome innate immune responses and concurrently uncover a mechanism that may exacerbate animal-borne MRSP infections in humans.
Collapse
Affiliation(s)
- Rita Haller
- Research Group Pathogenesis of Bacterial Infections; TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - Yiyang Cai
- Research Group Pathogenesis of Bacterial Infections; TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
- Department of Internal Medicine V, Universities of Giessen and Marburg Lung Center, University Hospital Giessen, Justus Liebig University Giessen, Member of the German Center for Lung Research (DZL), Giessen, Germany
- German Center for Infection Research (DZIF), Partner Site Giessen-Marburg-Langen, Justus Liebig University Giessen, Giessen, Germany
- Institute of Medical Microbiology, Justus Liebig University Giessen, Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany
- Institute for Lung Health (ILH), Justus Liebig University Giessen, Giessen, Germany
| | - Nicole de Buhr
- Institute of Biochemistry, University of Veterinary Medicine Hannover, Hannover, Germany
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Hannover, Germany
| | - Johanna C. Rieder
- Department of Small Animal Medicine and Surgery, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Dirk Schlüter
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence-Resolving Infection Susceptibility (RESIST), (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Claas Baier
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - Holger Rohde
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maren von Köckritz-Blickwede
- Institute of Biochemistry, University of Veterinary Medicine Hannover, Hannover, Germany
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Hannover, Germany
| | - Marius Vital
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - Volker Winstel
- Research Group Pathogenesis of Bacterial Infections; TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
- Department of Internal Medicine V, Universities of Giessen and Marburg Lung Center, University Hospital Giessen, Justus Liebig University Giessen, Member of the German Center for Lung Research (DZL), Giessen, Germany
- German Center for Infection Research (DZIF), Partner Site Giessen-Marburg-Langen, Justus Liebig University Giessen, Giessen, Germany
- Institute of Medical Microbiology, Justus Liebig University Giessen, Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany
- Institute for Lung Health (ILH), Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
38
|
Li A, Yi Z, Ma C, Sun B, Zhao L, Cheng X, Hui L, Xia Y. Innate immune recognition in hepatitis B virus infection. Virulence 2025; 16:2492371. [PMID: 40253712 PMCID: PMC12013422 DOI: 10.1080/21505594.2025.2492371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 03/19/2025] [Accepted: 04/02/2025] [Indexed: 04/22/2025] Open
Abstract
Hepatitis B virus (HBV) remains a major global public health challenge, with approximately 254 million individuals chronically infected worldwide. The interaction between HBV and the innate immune system has garnered significant attention within the scientific community, with numerous studies exploring this relationship over the past several decades. While some research suggests that HBV infection activates the host's innate immune response, other studies indicate that HBV suppresses innate immune signaling pathways. These conflicting findings underscore the complexity of the HBV-innate immunity interaction, which remains inadequately understood. This review aims to clarify this interplay by examining it from three perspectives: (a) studies showing HBV activation of innate immunity; (b) evidence suggesting HBV suppression of innate immunity; and (c) findings that support HBV's role as a stealth virus. By synthesizing these perspectives, we aim to deepen the understanding of virus-host interactions that are crucial to HBV persistence and immune evasion, with potential implications for developing new therapeutic strategies for chronic HBV infection.
Collapse
Affiliation(s)
- Aixin Li
- School of Laboratory Medicine, Shandong Second Medical University, Weifang, China
| | - Zhengjun Yi
- School of Laboratory Medicine, Shandong Second Medical University, Weifang, China
| | - Chunqiang Ma
- School of Laboratory Medicine, Shandong Second Medical University, Weifang, China
| | - Bangyao Sun
- School of Laboratory Medicine, Shandong Second Medical University, Weifang, China
| | - Li Zhao
- School of Laboratory Medicine, Shandong Second Medical University, Weifang, China
| | - Xiaoming Cheng
- State Key Laboratory of Virology and Biosafety, Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Medical School, Wuhan University, Wuhan, China
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Jiangxia Laboratory, Wuhan, China
- Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, China
| | - Lixia Hui
- School of Laboratory Medicine, Shandong Second Medical University, Weifang, China
| | - Yuchen Xia
- State Key Laboratory of Virology and Biosafety, Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Medical School, Wuhan University, Wuhan, China
- Hubei Jiangxia Laboratory, Wuhan, China
- Pingyuan Laboratory, Henan, China
| |
Collapse
|
39
|
Qing J, Li C, Jiao N. Deciphering the causal link between gut microbiota and membranous nephropathy: insights into potential inflammatory mechanisms. Ren Fail 2025; 47:2476053. [PMID: 40083050 PMCID: PMC11912295 DOI: 10.1080/0886022x.2025.2476053] [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: 12/29/2024] [Revised: 02/23/2025] [Accepted: 02/24/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Membranous nephropathy (MN), a leading cause of adult nephrotic syndrome and renal failure, has been linked to gut microbiota (GM) and their metabolites. However, direct causal relationships and therapeutic implications remain unclear. METHODS We utilized a comprehensive GWAS dataset that encompasses GM, metabolites, and MN through two-sample Mendelian randomization (MR) analyses, bidirectional MR evaluations, and detailed sensitivity tests. RESULTS We identified strong causal associations between nine specific types of GM, including class Clostridia (OR = 1.816, 95%CI: 1.021-3.236, p = .042), class Melainabacteria (OR = 0.661, 95%CI: 0.439-0.996, p = .048), order Gastranaerophilales (OR = 0.689, 95%CI: 0.480-0.996, p = .044), genus Alistipes (OR = 0.480, 95%CI: 0.223-0.998, p = .049), genus Butyricicoccus (OR = 0.464, 95%CI: 0.216-0.995, p = .048), genus Butyrivibrio (OR = 0.799, 95%CI: 0.639-0.998, p = .048), genus Ruminococcaceae UCG003 (OR = 0.563, 95%CI: 0.362-0.877, p = .011), genus Streptococcus (OR = 0.619, 95%CI: 0.393-0.973, p = .038), and genus Oscillibacter (OR = 1.90, 95%CI: 1.06-3.40, p = .031). Additionally, the metabolite tryptophan also exhibited a significant causal influence on MN (OR = 0.852, 95%CI: 0.754-0.963, p = .010). Sensitivity and reverse MR analyses confirmed the robustness of these findings. Further exploration using gutMGene database suggests that GM may influence MN by affecting the release of inflammatory factors and modulating inflammatory pathways. CONCLUSION This study offers a comprehensive understanding of the causal links between GM, their metabolites, and MN, which highlight potential pathways for developing new preventive and therapeutic strategies for this condition.
Collapse
Affiliation(s)
- Jianbo Qing
- Department of Nephrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Nephrology, Shanxi Provincial People’s Hospital, Shanxi Medical University, Taiyuan, China
- Department of Nephrology, The Fifth Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Changqun Li
- Department of Nephrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Nan Jiao
- Department of Nephrology, Shanxi Provincial People’s Hospital, Shanxi Medical University, Taiyuan, China
- Department of Nephrology, The Fifth Clinical Medical College, Shanxi Medical University, Taiyuan, China
- Big Data Center of Kidney Disease, Shanxi Provincial People’s Hospital, Taiyuan, China
- Shanxi Provincial Key Laboratory of Kidney Disease, Taiyuan, China
| |
Collapse
|
40
|
Fines C, McCarthy H, Buckley N. The search for a TNBC vaccine: the guardian vaccine. Cancer Biol Ther 2025; 26:2472432. [PMID: 40089851 PMCID: PMC11913391 DOI: 10.1080/15384047.2025.2472432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/17/2025] Open
Abstract
Nearly 20 million people are diagnosed with cancer each year with breast cancer being the most common among women. Triple negative breast cancer (TNBC), defined by its no/low expression of ER and PR and lack of amplification of HER2, makes up 15-20% of all breast cancer cases. While patients overall have a higher response to chemotherapy, this subgroup is associated with the lowest survival rate indicating significant clinical and molecular heterogeneity demanding alternate treatment options. Therefore, new therapies have been explored, with a large focus on utilizing the immune system. A whole host of immunotherapies have been studied including immune checkpoint inhibitors, now standard of care for eligible patients, and possibly the most exciting and promising is that of a TNBC vaccine. While currently there are no approved TNBC vaccines, this review highlights many promising studies and points to an antigen, p53, which we believe is highly relevant for TNBC.
Collapse
Affiliation(s)
- Cory Fines
- School of Pharmacy, Queen’s University Belfast, Belfast, UK
| | - Helen McCarthy
- School of Pharmacy, Queen’s University Belfast, Belfast, UK
| | - Niamh Buckley
- School of Pharmacy, Queen’s University Belfast, Belfast, UK
| |
Collapse
|
41
|
Liang Y, Du M, Li X, Gao J, Li Q, Li H, Li J, Gao X, Cong H, Huang Y, Li X, Wang L, Cui J, Gan Y, Tu H. Upregulation of Lactobacillus spp. in gut microbiota as a novel mechanism for environmental eustress-induced anti-pancreatic cancer effects. Gut Microbes 2025; 17:2470372. [PMID: 39988618 PMCID: PMC11853549 DOI: 10.1080/19490976.2025.2470372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 12/01/2024] [Accepted: 02/17/2025] [Indexed: 02/25/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignancy with limited effective treatment options. Emerging evidence links enriched environment (EE)-induced eustress to PDAC inhibition. However, the underlying mechanisms remain unclear. In this study, we explored the role of gut microbiota in PDAC-suppressive effects of EE. We demonstrated that depletion of gut microbiota with antibiotics abolished EE-induced tumor suppression, while fecal microbiota transplantation (FMT) from EE mice significantly inhibited tumor growth in both subcutaneous and orthotopic PDAC models housed in standard environment. 16S rRNA sequencing revealed that EE enhanced gut microbiota diversity and selectively enriched probiotic Lactobacillus, particularly L. reuteri. Treatment with L. reuteri significantly suppressed PDAC tumor growth and increased natural killer (NK) cell infiltration into the tumor microenvironment. Depletion of NK cells alleviated the anti-tumor effects of L. reuteri, underscoring the essential role of NK cell-mediated immunity in anti-tumor response. Clinical analysis of PDAC patients showed that higher fecal Lactobacillus abundance correlated with improved progression-free and overall survival, further supporting the therapeutic potential of L. reuteri in PDAC. Overall, this study identifies gut microbiota as a systemic regulator of PDAC under psychological stress. Supplementation of psychobiotic Lactobacillus may offer a novel therapeutic strategy for PDAC.
Collapse
Affiliation(s)
- Yiyi Liang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Du
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Gao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huimin Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jin Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiang Gao
- School of Basic Medicine, Fudan University, Shanghai, China
| | - Hui Cong
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yimeng Huang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinran Li
- School of Basic Medicine, Fudan University, Shanghai, China
| | - Liwei Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiujie Cui
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Gan
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Tu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
42
|
Xie Y, Mi X, Xing Y, Dai Z, Pu Q. Past, present, and future of exosomes research in cancer: A bibliometric and visualization analysis. Hum Vaccin Immunother 2025; 21:2488551. [PMID: 40207548 PMCID: PMC11988232 DOI: 10.1080/21645515.2025.2488551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/12/2025] [Accepted: 04/01/2025] [Indexed: 04/11/2025] Open
Abstract
Cancer seriously threatens the lives and health of people worldwide, and exosomes seem to play an important role in managing cancer effectively, which has attracted extensive attention from researchers in recent years. This study aimed to scientifically visualize exosomes research in cancer (ERC) through bibliometric analysis, reviewing the past, summarizing the present, and predicting the future, with a view to providing valuable insights for scholars and policy makers. Researches search and data collection from Web of Science Core Collection and clinical trial.gov. Calculations and visualizations were performed using Microsoft Excel, VOSviewer, Bibliometrix R-package, and CiteSpace. As of December 1, 2024, and March 8, 2025, we identified 8,001 ERC-related publications and 107 ERC-related clinical trials, with an increasing trend in annual publications. Our findings supported that China, Nanjing Medical University, and International Journal of Molecular Sciences were the most productive countries, institutions, and journals, respectively. Whiteside, Theresa L. had the most publications, while Théry, C was the most co-cited scholar. In addition, Cancer Research was the most co-cited journal. Spatial and temporal distribution of clinical trials was the same as for publications. High-frequency keywords were "extracellular vesicle," "microRNA" and "biomarker." Additional, "surface functionalization," "plant," "machine learning," "nanomaterials," "promotes metastasis," "engineered exosomes," and "macrophage-derived exosomes" were promising research topics. Our study comprehensively and visually summarized the structure, hotspots, and evolutionary trends of ERC. It would inspire subsequent studies from a macroscopic perspective and provide a basis for rational allocation of resources and identification of collaborations among researchers.
Collapse
Affiliation(s)
- Yafei Xie
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Xingqi Mi
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Yikai Xing
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Zhangyi Dai
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Qiang Pu
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
43
|
Long Z, Yi Z, Yan W, Wang H. Trends in the immunotherapy for glioblastoma: A two-decade bibliometric analysis. Hum Vaccin Immunother 2025; 21:2466299. [PMID: 39950580 PMCID: PMC11834472 DOI: 10.1080/21645515.2025.2466299] [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: 08/30/2024] [Revised: 01/27/2025] [Accepted: 02/09/2025] [Indexed: 02/20/2025] Open
Abstract
Glioblastoma is a life-threatening primary malignant brain tumor with an unfavorable prognosis. Contributing factors to its poor outcome include tumor heterogeneity, low mutational burden, and immunosuppression within the tumor microenvironment. Recognizing these challenges, immunotherapeutic strategies have emerged as a promising avenue for glioblastoma treatment. Although several dynamic research and scientific trend have increasingly taken pace in the immunotherapeutic approaches to glioblastoma, systematic bibliometric studies on such trends are few. On this note, this study explores a bibliometric analysis of the research hotspots and trends in glioblastoma immunotherapy. We conducted a search in the Web of Science Core Collection database for articles on glioblastoma immunotherapy published between 2004 and 2024. Using VOSviewer and CiteSpace software, we analyzed collected articles to explore aspects such as country of origin, journal of publication, affiliated institute, authorship, keywords, and citation patterns. As of May 1, 2024, we retrieved 3,729 papers on Glioblastoma Immunotherapy. In the field of glioblastoma immunotherapy, the United States stands out as the leading contributor, with 1,708 publications and a substantial 90,590 citations. Following closely, China has made significant contributions through 926 publications, earning 17,533 citations, while Germany adds to the body of knowledge with 349 publications and 16,355 citations. Furthermore, Authoritative journals in this field include Clinical Cancer Research and Neuro-Oncology. The top five keywords during this period were temozolomide, radiotherapy, dendritic cell, cytotoxic T lymphocyte, and vaccination. Moreover, Hotspots in the field include immune checkpoint inhibitors and chimeric antigen receptor T cell therapy.
Collapse
Affiliation(s)
- Zhi Long
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hypothalamic-Pituitary Research Center, 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
- Hypothalamic-Pituitary Research Center, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Yan
- The First Department of General Surgery, Hunan Provincial People’s Hospital, Hunan Normal University, Changsha, China
| | - Hongxin Wang
- Department of Neurosurgery, The Affiliated Changsha Central Hospital, Hengyang Medical School,University of South China, Changsha, China
| |
Collapse
|
44
|
Shang C, Shi S, Jiang Q, Wang X, Yao X, Li W, Song G, Li Y, Sun Y, Hu J, Zhang C, Zhu Y, Liu Z, Gu C, Liu Y, Shi W, Zhao Z, Li X. Clinical manifestations and pathogenicity of Clade IIb monkeypox virus in rabbits. Emerg Microbes Infect 2025; 14:2465309. [PMID: 39945750 PMCID: PMC11849023 DOI: 10.1080/22221751.2025.2465309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 01/29/2025] [Accepted: 02/05/2025] [Indexed: 02/18/2025]
Abstract
The 2022 monkeypox outbreak involved rapid global dissemination, prompting research into animal models for the monkeypox virus (MPXV), including non-human primates and mice. However, studies utilizing rabbits as models remain limited. In this study, we established three rabbit models using the current epidemic MPXV strain. Following intravenous MPXV injection, adult rabbits exhibited characteristic clinical manifestations, including widespread rash and fever, with viral replication in the skin, lungs, and testes, resulting in severe pathological damage by 6 days post-infection (dpi). Intradermal injection of MPXV into the dorsal skin of adult rabbits produced red lesions with central necrosis and hemorrhage accompanied by dense inflammatory infiltrates. Abundant viral particles were observed in epidermal cells at 6 dpi. Additionally, a fatal MPXV model was developed in 10-day-old rabbits using intranasal virus administration. These young rabbits exhibited lethargy and diarrhea beginning at 2 dpi, significant weight loss, and a 50% mortality rate by 15 dpi. Viral dissemination was detected in multiple organs, leading to extensive multi-organ damage. This study highlights the utility of rabbit models for MPXV, displaying typical clinical features and pathogenic mechanisms.
Collapse
Affiliation(s)
- Chao Shang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Shaowen Shi
- Hebei Agricultural University, Baoding, People’s Republic of China
| | - Qiwei Jiang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Xiaohan Wang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Xiaohong Yao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, People’s Republic of China
| | - Wanzi Li
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, People’s Republic of China
| | - Gaojie Song
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang, People’s Republic of China
| | - Yiquan Li
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Yongyang Sun
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Jinglei Hu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Cuiling Zhang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Yilong Zhu
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Zirui Liu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Chaode Gu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Yan Liu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Wanyu Shi
- Hebei Agricultural University, Baoding, People’s Republic of China
| | - Zongzheng Zhao
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Xiao Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| |
Collapse
|
45
|
Castagnino PA, Haas DA, Musante L, Tancler NA, Tran BV, Kean R, Steck AR, Martinez LA, Mostaghel EA, Hooper DC, Kim FJ. Sigma1 inhibitor suppression of adaptive immune resistance mechanisms mediated by cancer cell derived extracellular vesicles. Cancer Biol Ther 2025; 26:2455722. [PMID: 39863992 PMCID: PMC11776462 DOI: 10.1080/15384047.2025.2455722] [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: 08/07/2024] [Revised: 12/28/2024] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Adaptive immune resistance in cancer describes the various mechanisms by which tumors adapt to evade anti-tumor immune responses. IFN-γ induction of programmed death-ligand 1 (PD-L1) was the first defined and validated adaptive immune resistance mechanism. The endoplasmic reticulum (ER) is central to adaptive immune resistance as immune modulatory secreted and integral membrane proteins are dependent on ER. Sigma1 is a unique ligand-regulated integral membrane scaffolding protein enriched in the ER of cancer cells. PD-L1 is an integral membrane glycoprotein that is translated into the ER and processed through the cellular secretory pathway. At the cell surface, PD-L1 is an immune checkpoint molecule that binds PD-1 on activated T-cells and blocks anti-tumor immunity. PD-L1 can also be incorporated into cancer cell-derived extracellular vesicles (EVs), and EV-associated PD-L1 can inactivate T-cells within the tumor microenvironment. Here, we demonstrate that a selective small molecule inhibitor of Sigma1 can block IFN-γ mediated adaptive immune resistance in part by altering the incorporation of PD-L1 into cancer cell-derived EVs. Sigma1 inhibition blocked post-translational maturation of PD-L1 downstream of IFN-γ/STAT1 signaling. Subsequently, EVs released in response to IFN-γ stimulation were significantly less potent suppressors of T-cell activation. These results suggest that by reducing tumor derived immune suppressive EVs, Sigma1 inhibition may promote antitumor immunity. Sigma1 modulation presents a novel approach to regulating the tumor immune microenvironment by altering the content and production of EVs. Altogether, these data support the notion that Sigma1 may play a role in adaptive immune resistance in the tumor microenvironment.
Collapse
Affiliation(s)
- Paola A. Castagnino
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Comprehensive Cancer Center at Jefferson, Philadelphia, PA, USA
| | - Derick A. Haas
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Comprehensive Cancer Center at Jefferson, Philadelphia, PA, USA
| | - Luca Musante
- University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | - Nathalia A. Tancler
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Comprehensive Cancer Center at Jefferson, Philadelphia, PA, USA
| | - Bach V. Tran
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Comprehensive Cancer Center at Jefferson, Philadelphia, PA, USA
| | - Rhonda Kean
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Comprehensive Cancer Center at Jefferson, Philadelphia, PA, USA
| | - Alexandra R. Steck
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Comprehensive Cancer Center at Jefferson, Philadelphia, PA, USA
| | - Luis A. Martinez
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Comprehensive Cancer Center at Jefferson, Philadelphia, PA, USA
| | - Elahe A. Mostaghel
- Geriatric Research, Education and Clinical Center, U.S. Department of Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
| | - D. Craig Hooper
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Comprehensive Cancer Center at Jefferson, Philadelphia, PA, USA
| | - Felix J. Kim
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Comprehensive Cancer Center at Jefferson, Philadelphia, PA, USA
| |
Collapse
|
46
|
Rodrigues CS, Gaifem J, Pereira MS, Alves MF, Silva M, Padrão N, Cavadas B, Moreira-Barbosa C, Alves I, Marcos-Pinto R, Torres J, Lavelle A, Colombel JF, Sokol H, Pinho SS. Alterations in mucosa branched N-glycans lead to dysbiosis and downregulation of ILC3: a key driver of intestinal inflammation. Gut Microbes 2025; 17:2461210. [PMID: 39918275 PMCID: PMC11810091 DOI: 10.1080/19490976.2025.2461210] [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: 08/29/2024] [Revised: 11/29/2024] [Accepted: 01/13/2025] [Indexed: 02/12/2025] Open
Abstract
The perturbation of the symbiotic relationship between microbes and intestinal immune system contributes to gut inflammation and Inflammatory Bowel Disease (IBD) development. The host mucosa glycans (glycocalyx) creates a major biological interface between gut microorganisms and host immunity that remains ill-defined. Glycans are essential players in IBD immunopathogenesis, even years before disease onset. However, how changes in mucosa glycosylation shape microbiome and how this impact gut immune response and inflammation remains to be clarified. Here, we revealed that alterations in the expression of complex branched N-glycans at gut mucosa surface, modeled in glycoengineered mice, resulted in dysbiosis, with a deficiency in Firmicutes bacteria. Concomitantly, this mucosa N-glycan switch was associated with a downregulation of type 3 innate lymphoid cells (ILC3)-mediated immune response, leading to the transition of ILC3 toward an ILC1 proinflammatory phenotype and increased TNFα production. In addition, we demonstrated that the mucosa glycosylation remodeling through prophylactic supplementation with glycans at steady state was able to restore microbial-derived short-chain fatty acids and microbial sensing (by NOD2 expression) alongside the rescue of the expression of ILC3 module, suppressing intestinal inflammation and controlling disease onset. In a complementary approach, we further showed that IBD patients, often displaying dysbiosis, exhibited a tendency of decreased MGAT5 expression at epithelial cells that was accompanied by reduced ILC3 expression in gut mucosa. Altogether, these results unlock the effects of alterations in mucosa glycome composition in the regulation of the bidirectional crosstalk between microbiota and gut immune response, revealing host branched N-glycans/microbiota/ILC3 axis as an essential pathway in gut homeostasis and in preventing health to intestinal inflammation transition.
Collapse
Affiliation(s)
- Cláudia S. Rodrigues
- Institute for Research and Innovation in Health (i3S), Immunology, Cancer & Glycomedicine Group, University of Porto, Porto, Portugal
- School of Medicine and Biomedical Sciences (ICBAS), University of Porto, Porto, Portugal
| | - Joana Gaifem
- Institute for Research and Innovation in Health (i3S), Immunology, Cancer & Glycomedicine Group, University of Porto, Porto, Portugal
| | - Márcia S. Pereira
- Institute for Research and Innovation in Health (i3S), Immunology, Cancer & Glycomedicine Group, University of Porto, Porto, Portugal
- School of Medicine and Biomedical Sciences (ICBAS), University of Porto, Porto, Portugal
| | - Maria Francisca Alves
- Institute for Research and Innovation in Health (i3S), Immunology, Cancer & Glycomedicine Group, University of Porto, Porto, Portugal
- School of Medicine and Biomedical Sciences (ICBAS), University of Porto, Porto, Portugal
- Faculty of Sciences, University of Porto, Porto, Portugal
| | - Mariana Silva
- Institute for Research and Innovation in Health (i3S), Immunology, Cancer & Glycomedicine Group, University of Porto, Porto, Portugal
- School of Medicine and Biomedical Sciences (ICBAS), University of Porto, Porto, Portugal
| | - Nuno Padrão
- Institute for Research and Innovation in Health (i3S), Immunology, Cancer & Glycomedicine Group, University of Porto, Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
| | - Bruno Cavadas
- Institute for Research and Innovation in Health (i3S), Immunology, Cancer & Glycomedicine Group, University of Porto, Porto, Portugal
| | | | - Inês Alves
- Institute for Research and Innovation in Health (i3S), Immunology, Cancer & Glycomedicine Group, University of Porto, Porto, Portugal
| | - Ricardo Marcos-Pinto
- School of Medicine and Biomedical Sciences (ICBAS), University of Porto, Porto, Portugal
- Department of Gastroenterology, Centro Hospitalar do Porto, Porto, Portugal
- Centro de Investigação em Tecnologias e Serviços de Saúde, University of Porto, Porto, Portugal
| | - Joana Torres
- Division of Gastroenterology, Hospital Beatriz Ângelo, Loures, Portugal
- Faculty of Medicine, University of Lisbon, Lisbon, Portugal
- Division of Gastroenterology, Hospital da Luz, Lisbon, Portugal
| | - Aonghus Lavelle
- Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint-Antoine Hospital, Gastroenterology Department, Sorbonne Université, INSERM, Paris, France
| | - Jean-Frederic Colombel
- Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Harry Sokol
- Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint-Antoine Hospital, Gastroenterology Department, Sorbonne Université, INSERM, Paris, France
- INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, Jouy-en-Josas, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Salomé S. Pinho
- Institute for Research and Innovation in Health (i3S), Immunology, Cancer & Glycomedicine Group, University of Porto, Porto, Portugal
- School of Medicine and Biomedical Sciences (ICBAS), University of Porto, Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
47
|
Guo F, Song Y, Dong S, Wei J, Li B, Xu T, Wang H. Characterization and anti-tuberculosis effects of γδ T cells expanded and activated by Mycobacterium tuberculosis heat-resistant antigen. Virulence 2025; 16:2462092. [PMID: 39921673 PMCID: PMC11810100 DOI: 10.1080/21505594.2025.2462092] [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: 08/09/2024] [Revised: 12/01/2024] [Accepted: 01/20/2025] [Indexed: 02/10/2025] Open
Abstract
Tuberculosis (TB) is a chronic infectious disease caused by Mycobacterium tuberculosis (Mtb) that poses a severe threat to human health. A variety of highly immunogenic tuberculosis proteins have been used as targets in vaccine development to mitigate the spread of TB. Although Th1-type immunity has long been considered a crucial part of resistance to Mtb, γδ T cells, the predominant source of IL-17, are not negligible in controlling the early stages of TB infection. In addition to classical phosphoantigens, Mycobacterium tuberculosis heat-resistant antigens (HAg), a complex containing 564 proteins obtained from live tuberculosis bacteria after heat treatment at 121 °C for 20 min, have been confirmed to be highly effective γδ T cell stimulators as well. Several studies have demonstrated that HAg-activated γδ T cells can participate in TB immunity by secreting multiple cytokines against Mtb or by interacting with other innate immune cells. In this review, we present a possible mechanism of HAg stimulation of γδ T cells and the role of HAg-activated γδ T cells in anti-TB immunity. We also highlight the limitations of studies on HAg activation of γδ T cells and suggest further research directions on the relationship between HAg and γδ T cells.
Collapse
Affiliation(s)
- Fangzheng Guo
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Research Center of Laboratory, School of Laboratory, Bengbu Medical University, Bengbu, China
- Department of Immunology, School of Laboratory, Bengbu Medical University, Bengbu, China
| | - Yamin Song
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Research Center of Laboratory, School of Laboratory, Bengbu Medical University, Bengbu, China
- Department of Immunology, School of Laboratory, Bengbu Medical University, Bengbu, China
| | - Sihang Dong
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Research Center of Laboratory, School of Laboratory, Bengbu Medical University, Bengbu, China
- Department of Immunology, School of Laboratory, Bengbu Medical University, Bengbu, China
| | - Jing Wei
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Research Center of Laboratory, School of Laboratory, Bengbu Medical University, Bengbu, China
- Department of Immunology, School of Laboratory, Bengbu Medical University, Bengbu, China
| | - Baiqing Li
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Research Center of Laboratory, School of Laboratory, Bengbu Medical University, Bengbu, China
- Department of Immunology, School of Laboratory, Bengbu Medical University, Bengbu, China
- Department of Clinical Laboratory, School of Laboratory, Bengbu Medical University, Bengbu, China
| | - Tao Xu
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Research Center of Laboratory, School of Laboratory, Bengbu Medical University, Bengbu, China
- Department of Immunology, School of Laboratory, Bengbu Medical University, Bengbu, China
- Department of Clinical Laboratory, School of Laboratory, Bengbu Medical University, Bengbu, China
| | - Hongtao Wang
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Research Center of Laboratory, School of Laboratory, Bengbu Medical University, Bengbu, China
- Department of Immunology, School of Laboratory, Bengbu Medical University, Bengbu, China
- Department of Clinical Laboratory, School of Laboratory, Bengbu Medical University, Bengbu, China
| |
Collapse
|
48
|
Lin Y, Wang J, Bu F, Zhang R, Wang J, Wang Y, Huang M, Huang Y, Zheng L, Wang Q, Hu X. Bacterial extracellular vesicles in the initiation, progression and treatment of atherosclerosis. Gut Microbes 2025; 17:2452229. [PMID: 39840620 DOI: 10.1080/19490976.2025.2452229] [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: 08/21/2024] [Revised: 11/13/2024] [Accepted: 01/07/2025] [Indexed: 01/23/2025] Open
Abstract
Atherosclerosis is the primary cause of cardiovascular and cerebrovascular diseases. However, current anti-atherosclerosis drugs have shown conflicting therapeutic outcomes, thereby spurring the search for novel and effective treatments. Recent research indicates the crucial involvement of oral and gastrointestinal microbiota in atherosclerosis. While gut microbiota metabolites, such as choline derivatives, have been extensively studied and reviewed, emerging evidence suggests that bacterial extracellular vesicles (BEVs), which are membrane-derived lipid bilayers secreted by bacteria, also play a significant role in this process. However, the role of BEVs in host-microbiota interactions remains insufficiently explored. This review aims to elucidate the complex communication mediated by BEVs along the gut-heart axis. In this review, we summarize current knowledge on BEVs, with a specific focus on how pathogen-derived BEVs contribute to the promotion of atherosclerosis, as well as how BEVs from gut symbionts and probiotics may mitigate its progression. We also explore the potential and challenges associated with engineered BEVs in the prevention and treatment of atherosclerosis. Finally, we discuss the benefits and challenges of using BEVs in atherosclerosis diagnosis and treatment, and propose future research directions to address these issues.
Collapse
Affiliation(s)
- Yuling Lin
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jingyu Wang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fan Bu
- Institute of Hematology, Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Zhejiang University, Hangzhou, China
| | - Ruyi Zhang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Junhui Wang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yubing Wang
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Mei Huang
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yiyi Huang
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qian Wang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiumei Hu
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
49
|
Rosati D, Valentine M, Bruno M, Pradhan A, Dietschmann A, Jaeger M, Leaves I, van de Veerdonk FL, Joosten LA, Roy S, Stappers MHT, Gow NA, Hube B, Brown AJ, Gresnigt MS, Netea MG. Lactic acid in the vaginal milieu modulates the Candida-host interaction. Virulence 2025; 16:2451165. [PMID: 39843417 PMCID: PMC11760238 DOI: 10.1080/21505594.2025.2451165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/07/2024] [Accepted: 12/28/2024] [Indexed: 01/24/2025] Open
Abstract
Vulvovaginal candidiasis (VVC) is one of the most common infections caused by Candida albicans. VVC is characterized by an inadequate hyperinflammatory response and clinical symptoms associated with Candida colonization of the vaginal mucosa. Compared to other host niches in which C. albicans can cause infection, the vaginal environment is extremely rich in lactic acid that is produced by the vaginal microbiota. We examined how lactic acid abundance in the vaginal niche impacts the interaction between C. albicans and the human immune system using an in vitro culture in vaginal simulative medium (VSM). The presence of lactic acid in VSM (VSM+LA) increased C. albicans proliferation, hyphal length, and its ability to cause damage during subsequent infection of vaginal epithelial cells. The cell wall of C. albicans cells grown in VSM+LA displayed a robust mannan fibrillar structure, β-glucan exposure, and low chitin content. These cell wall changes were associated with altered immune responses and an increased ability of the fungus to induce trained immunity. Neutrophils were compromised in clearing C. albicans grown in VSM+LA conditions, despite mounting stronger oxidative responses. Collectively, we found that fungal adaptation to lactic acid in a vaginal simulative context increases its immunogenicity favouring a pro-inflammatory state. This potentially contributes to the immune response dysregulation and neutrophil recruitment observed during recurrent VVC.
Collapse
Affiliation(s)
- Diletta Rosati
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, T
he Netherlands
| | - Marisa Valentine
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans-Knöll-Institute, Jena, Germany
| | - Mariolina Bruno
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, T
he Netherlands
| | - Arnab Pradhan
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Axel Dietschmann
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology - Hans-Knöll-Institute, Jena, Germany
| | - Martin Jaeger
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, T
he Netherlands
| | - Ian Leaves
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Frank L. van de Veerdonk
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, T
he Netherlands
| | - Leo A.B. Joosten
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, T
he Netherlands
- Department of Medical Genetics, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Sumita Roy
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Mark H. T. Stappers
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Neil A.R. Gow
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans-Knöll-Institute, Jena, Germany
- Institute of Microbiology, Friedrich-Schiller-University, Jena, Germany
| | - Alistair J.P. Brown
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Mark S. Gresnigt
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology - Hans-Knöll-Institute, Jena, Germany
| | - Mihai G. Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, T
he Netherlands
- Department of Immunology and Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| |
Collapse
|
50
|
Li C, Ji H, Zhuang S, Xie X, Cui D, Zhang C. Update on the correlation between mitochondrial function and osteonecrosis of the femoral head osteocytes. Redox Rep 2025; 30:2491846. [PMID: 40249372 PMCID: PMC12010656 DOI: 10.1080/13510002.2025.2491846] [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: 04/19/2025] Open
Abstract
Mitochondrial health is maintained in a steady state through mitochondrial dynamics and autophagy processes. Recent studies have identified healthy mitochondria as crucial regulators of cellular function and survival. This process involves adenosine triphosphate (ATP) synthesis by mitochondrial oxidative phosphorylation (OXPHOS), regulation of calcium metabolism and inflammatory responses, and intracellular oxidative stress management. In the skeletal system, they participate in the regulation of cellular behaviors and the responses of osteoblasts, osteoclasts, chondrocytes, and osteocytes to external stimuli. Indeed, mitochondrial damage or dysfunction occurs in the development of a few bone diseases. For example, mitochondrial damage may lead to an imbalance in osteoblasts and osteoclasts, resulting in osteoporosis, osteomalacia, or poor bone production, and chondrocyte death and inflammatory infiltration in osteoarthritis are the main causes of cartilage degeneration due to mitochondrial damage. However, the opposite exists for osteosarcoma, where overactive mitochondrial metabolism is able to accelerate the proliferation and migration of osteosarcoma cells, which is a major disease feature. Bone is a dynamic organ and osteocytes play a fundamental role in all regions of bone tissue and are involved in regulating bone integrity. This review examines the impact of mitochondrial physiological function on osteocyte health and summarizes the microscopic molecular mechanisms underlying its effects. It highlights that targeted therapies focusing on osteocyte mitochondria may be beneficial for osteocyte survival, providing a new insight for the diagnosis, prevention, and treatment of diseases associated with osteocyte death.
Collapse
Affiliation(s)
- Chengming Li
- Department of Orthopedics, Zhongda Hospital Southeast University, Nanjing, People’s Republic of China
| | - Hangyu Ji
- Department of Orthopedics, Zhongda Hospital Southeast University, Nanjing, People’s Republic of China
| | - Suyang Zhuang
- Department of Orthopedics, Zhongda Hospital Southeast University, Nanjing, People’s Republic of China
| | - Xinhui Xie
- Department of Orthopedics, Zhongda Hospital Southeast University, Nanjing, People’s Republic of China
| | - Daping Cui
- Department of Orthopedics, Shenzhen Bao’an District Central Hospital, Shenzhen, People’s Republic of China
| | - Cong Zhang
- Department of Orthopedics, Zhongda Hospital Southeast University, Nanjing, People’s Republic of China
| |
Collapse
|