1
|
He P, He C, Guo R, Ou Y, Chang Y, Xie Z, Tang X, Xu Y, Zhao Y, Wang H, Guo Z, Bai S, Chen Z, Fan F, Du G, Sun X. Tough and waterproof microneedles overcome mucosal immunotolerance by modulating antigen release patterns. J Control Release 2025; 382:113740. [PMID: 40250628 DOI: 10.1016/j.jconrel.2025.113740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 04/02/2025] [Accepted: 04/14/2025] [Indexed: 04/20/2025]
Abstract
Mucosal vaccines are considered an ideal choice for combating mucosal pathogens due to their ability to neutralize pathogens at the first line of defense. However, the development of mucosal subunit vaccines is constricted by rigorous challenges, such as low immunogenicity, poor antigen delivery efficiency, and mucosal tolerance. Here, a buccal microneedle patch incorporated with engineered nanoparticles loaded with urease B subunit (rUreB) was developed to overcome the above challenges. Specifically, an engineered nanocarrier was developed to protect the antigen and modulate its release profile. Then, the nanoparticles were enriched to form microneedle tips with superior mechanical and waterproof properties, allowing effective penetration of the buccal mucosa and resistance to salivary washout. Besides, the microneedles demonstrated an S-curve antigen release pattern which was crucial for the recruitment of antigen presenting cells (APCs) and the downregulation of mucosal tolerogenic DCs and Treg cells. The buccal microneedle vaccine without any immune stimulators induced potent systemic and mucosal immune responses, resulting in superior protection of mice from the oral challenge of Helicobacter pylori. These results suggested that the rationally designed buccal microneedle vaccine can effectively overcome mucosal delivery barriers and mucosal tolerance, providing a promising alternative strategy for mucosal vaccination of subunit antigens.
Collapse
Affiliation(s)
- Penghui He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Chunting He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Rong Guo
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yangsen Ou
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yu Chang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zhiqiang Xie
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xue Tang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yanhua Xu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yuanhao Zhao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Hairui Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zhaofei Guo
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Shuting Bai
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zhengjun Chen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Fan Fan
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Guangsheng Du
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xun Sun
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
2
|
Meden A, Claes S, Van Loy T, Zorman M, Proj M, Schols D, Gobec S, De Jonghe S. Structure-activity relationship study of navarixin analogues as dual CXCR2 and CCR7 antagonists. Bioorg Chem 2025; 159:108423. [PMID: 40179581 DOI: 10.1016/j.bioorg.2025.108423] [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: 11/14/2024] [Revised: 03/21/2025] [Accepted: 03/26/2025] [Indexed: 04/05/2025]
Abstract
Despite the promise of the human chemokine receptor 7 (CCR7) as drug target for the treatment of cancer metastasis and autoimmune diseases, there are no potent and selective CCR7 antagonists known in literature. In this work, a 1,2,5-thiadiazole 1,1-dioxide with low μM activity as a CXCR2 and CCR7 antagonist was selected as starting point for a structure-activity relationship study. The replacement of the central thiadiazole dioxide motif with squaramide led to low nanomolar CCR7 antagonism. Additional systematic structural variations afforded various squaramide analogues that displayed potent CCR7 antagonism in a calcium mobilization assay with IC50 values in the low nM range. Unfortunately, the same compounds also displayed potent CXCR2 antagonistic activity and should therefore be considered as dual CCR7/CXCR2 antagonists.
Collapse
Affiliation(s)
- Anže Meden
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Askerceva 7, 1000 Ljubljana, Slovenia
| | - Sandra Claes
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Molecular, Structural and Translational Virology Research Group, Herestraat 49, box 1043, 3000 Leuven, Belgium
| | - Tom Van Loy
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Molecular, Structural and Translational Virology Research Group, Herestraat 49, box 1043, 3000 Leuven, Belgium
| | - Maša Zorman
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Askerceva 7, 1000 Ljubljana, Slovenia
| | - Matic Proj
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Askerceva 7, 1000 Ljubljana, Slovenia
| | - Dominique Schols
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Molecular, Structural and Translational Virology Research Group, Herestraat 49, box 1043, 3000 Leuven, Belgium
| | - Stanislav Gobec
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Askerceva 7, 1000 Ljubljana, Slovenia.
| | - Steven De Jonghe
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Molecular, Structural and Translational Virology Research Group, Herestraat 49, box 1043, 3000 Leuven, Belgium.
| |
Collapse
|
3
|
Pentimalli TM, Schallenberg S, León-Periñán D, Legnini I, Theurillat I, Thomas G, Boltengagen A, Fritzsche S, Nimo J, Ruff L, Dernbach G, Jurmeister P, Murphy S, Gregory MT, Liang Y, Cordenonsi M, Piccolo S, Coscia F, Woehler A, Karaiskos N, Klauschen F, Rajewsky N. Combining spatial transcriptomics and ECM imaging in 3D for mapping cellular interactions in the tumor microenvironment. Cell Syst 2025; 16:101261. [PMID: 40220761 DOI: 10.1016/j.cels.2025.101261] [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: 07/12/2024] [Revised: 12/13/2024] [Accepted: 03/19/2025] [Indexed: 04/14/2025]
Abstract
Tumors are complex ecosystems composed of malignant and non-malignant cells embedded in a dynamic extracellular matrix (ECM). In the tumor microenvironment, molecular phenotypes are controlled by cell-cell and ECM interactions in 3D cellular neighborhoods (CNs). While their inhibition can impede tumor progression, routine molecular tumor profiling fails to capture cellular interactions. Single-cell spatial transcriptomics (ST) maps receptor-ligand interactions but usually remains limited to 2D tissue sections and lacks ECM readouts. Here, we integrate 3D ST with ECM imaging in serial sections from one clinical lung carcinoma to systematically quantify molecular states, cell-cell interactions, and ECM remodeling in CN. Our integrative analysis pinpointed known immune escape and tumor invasion mechanisms, revealing several druggable drivers of tumor progression in the patient under study. This proof-of-principle study highlights the potential of in-depth CN profiling in routine clinical samples to inform microenvironment-directed therapies. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- Tancredi Massimo Pentimalli
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany; Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin
| | - Simon Schallenberg
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Daniel León-Periñán
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Ivano Legnini
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany; Human Technopole, Milan, Italy
| | - Ilan Theurillat
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Gwendolin Thomas
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Anastasiya Boltengagen
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Sonja Fritzsche
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Spatial Proteomics Group, Berlin, Germany; Humboldt-Universität zu Berlin, Institute of Biology, 10099 Berlin, Germany
| | - Jose Nimo
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin; Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Spatial Proteomics Group, Berlin, Germany; Humboldt-Universität zu Berlin, Institute of Biology, 10099 Berlin, Germany
| | | | - Gabriel Dernbach
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany; Aignostics GmbH, Berlin, Germany; BIFOLD - Berlin Institute for the Foundations of Learning and Data, Berlin, Germany
| | | | | | | | - Yan Liang
- NanoString® Technologies, Inc, Seattle, WA, USA
| | | | - Stefano Piccolo
- Department of Molecular Medicine, University of Padua, Padua, Italy; IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Fabian Coscia
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Spatial Proteomics Group, Berlin, Germany
| | - Andrew Woehler
- Systems Biology Imaging Platform, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany; Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA, USA
| | - Nikos Karaiskos
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Frederick Klauschen
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin; BIFOLD - Berlin Institute for the Foundations of Learning and Data, Berlin, Germany; Institute of Pathology, Ludwig Maximilians Universität, Munich, Germany
| | - Nikolaus Rajewsky
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany; Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin; German Center for Cardiovascular Research (DZHK), Site Berlin, Berlin, Germany; NeuroCure Cluster of Excellence, Berlin, Germany; German Cancer Consortium (DKTK), Berlin, Germany; National Center for Tumor Diseases (NCT), Site Berlin, Berlin, Germany.
| |
Collapse
|
4
|
Chen X, Zheng Y, Man X, Li W. Tissue-resident memory T cells and their function in skin diseases. Chin Med J (Engl) 2025; 138:1175-1183. [PMID: 40066785 PMCID: PMC12091617 DOI: 10.1097/cm9.0000000000003499] [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: 06/21/2024] [Indexed: 05/21/2025] Open
Abstract
ABSTRACT Tissue-resident memory T (TRM) cells are a recently defined subtype of non-recirculating memory T cells with longevity and protective functions in peripheral tissues. As an essential frontline defense against infections, TRM cells have been reported to robustly patrol the tissue microenvironment in malignancies. Accumulating evidence also implicates that TRM cells in the relapse of chronic inflammatory skin diseases such as psoriasis and vitiligo. In light of these developments, this review aims to synthesize these recent findings to enhance our understanding of TRM cell characteristics and actions. Therefore, after providing a brief overview of the general features of the TRM cells, including precursors, homing, retention, and maintenance, we discuss recent insights gained into their heterogeneous functions in skin diseases. Specifically, we explore their involvement in conditions such as psoriasis, vitiligo, fixed drug eruption - dermatological manifestations of drug reactions at the same spot, cutaneous T cell lymphoma, and melanoma. By integrating these diverse perspectives, this review develops a comprehensive model of TRM cell behavior in various skin-related pathologies. In conclusion, our review emphasizes that deciphering the characteristics and mechanisms of TRM cell actions holds potential not only for discovering methods to slow cancer growth but also for reducing the frequency of recurrent chronic inflammation in skin tissue.
Collapse
Affiliation(s)
- Xibei Chen
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang 310009, China
| | - Yuxin Zheng
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang 310009, China
| | - Xiaoyong Man
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang 310009, China
| | - Wei Li
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang 310009, China
| |
Collapse
|
5
|
Wang M, Otto C, Fernández Zapata C, Dehlinger A, Gallaccio G, Diekmann LM, Niederschweiberer M, Schindler P, Körtvélyessy P, Kunkel D, Paul F, Ruprecht K, Böttcher C. Comprehensive analysis of B cell repopulation in ocrelizumab-treated patients with multiple sclerosis by mass cytometry and proteomics. iScience 2025; 28:112383. [PMID: 40322080 PMCID: PMC12049848 DOI: 10.1016/j.isci.2025.112383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 09/30/2024] [Accepted: 04/04/2025] [Indexed: 05/08/2025] Open
Abstract
Ocrelizumab, an anti-CD20 antibody, depletes CD20+ B cells, which subsequently repopulate over months. Little is known about changes in other immune cell populations and molecular markers associated with B cell repopulation. Here, we performed a comprehensive characterization of immune cells from ocrelizumab-treated patients with multiple sclerosis (MS) using mass cytometry. About 50% of patients showed naive B cell repopulation after 6 months mainly with a transitional phenotype, whereas CD27+ memory B cells only rarely repopulated. This repopulation was associated with a reduction of memory T cells and activated myeloid cells, as well as reduced expression of activation/migration markers in both cell types. A plasma proteomics analysis identified proteins including TNFRSF13C, associated with B cell depletion and repopulation. Plasma levels of neurofilament light-chain protein declined after ocrelizumab treatment was not linked with B cell repopulation. These findings identify potential soluble markers for monitoring of ocrelizumab treatment in MS.
Collapse
Affiliation(s)
- Meng Wang
- Experimental and Clinical Research Center, a Cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin Berlin, Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, Lindenberger Weg 80, Berlin 13125, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Carolin Otto
- Department of Neurology with Experimental Neurology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
| | - Camila Fernández Zapata
- Experimental and Clinical Research Center, a Cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin Berlin, Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, Lindenberger Weg 80, Berlin 13125, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Adeline Dehlinger
- Experimental and Clinical Research Center, a Cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin Berlin, Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, Lindenberger Weg 80, Berlin 13125, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Gerardina Gallaccio
- Experimental and Clinical Research Center, a Cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin Berlin, Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, Lindenberger Weg 80, Berlin 13125, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Lisa-Marie Diekmann
- Experimental and Clinical Research Center, a Cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin Berlin, Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, Lindenberger Weg 80, Berlin 13125, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Moritz Niederschweiberer
- Department of Neurology with Experimental Neurology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
| | - Patrick Schindler
- Experimental and Clinical Research Center, a Cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin Berlin, Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, Lindenberger Weg 80, Berlin 13125, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Department of Neurology with Experimental Neurology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
- Neuroscience Clinical Research Center, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
| | - Peter Körtvélyessy
- Department of Neurology, Center for ALS and Other Motor Neuron Disorders, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Desiree Kunkel
- Flow & MassCytometry Core Facility, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin 13353, Germany
| | - Friedemann Paul
- Experimental and Clinical Research Center, a Cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin Berlin, Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, Lindenberger Weg 80, Berlin 13125, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Department of Neurology with Experimental Neurology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
- Neuroscience Clinical Research Center, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
| | - Klemens Ruprecht
- Department of Neurology with Experimental Neurology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 10117, Germany
| | - Chotima Böttcher
- Experimental and Clinical Research Center, a Cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin Berlin, Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, Lindenberger Weg 80, Berlin 13125, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| |
Collapse
|
6
|
Pavan C, Davidson KC, Payne N, Frausin S, Hunt CPJ, Moriarty N, Berrocal Rubio MÁ, Elahi Z, Quattrocchi AT, Abu-Bonsrah KD, Wang L, Clow W, Yang H, Pellegrini M, Wells CA, Thompson LH, Nagy A, Parish CL. A cloaked human stem-cell-derived neural graft capable of functional integration and immune evasion in rodent models. Cell Stem Cell 2025; 32:710-726.e8. [PMID: 40209717 DOI: 10.1016/j.stem.2025.03.008] [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: 11/13/2024] [Accepted: 03/13/2025] [Indexed: 04/12/2025]
Abstract
Human pluripotent stem cell (hPSC)-derived therapies are a realistic possibility for numerous disorders, including Parkinson's disease. While generating replacement neurons is achievable, immunosuppressive drug challenges, to prevent rejection, remain. Here we adopted a hPSC line (termed H1-FS-8IM), engineered to overexpress 8 immunomodulatory transgenes, to enable transplant immune evasion. In co-cultures, H1-FS-8IM PSC-derived midbrain neurons evaded rejection by T lymphocytes, natural killer cells, macrophages, and dendritic cells. In humanized mice, allogeneic H1-FS-8IM neural grafts evaded rejection, while control hPSC-derived neural grafts evoked activation of human immune cells, elevated inflammatory cytokines in blood and cerebrospinal fluid, and caused spleen and lymph node enlargement. H1-FS-8IM neural grafts retained functionality, reversing motor deficits in Parkinsonian rats. Additional incorporation of a suicide gene into the H1-FS-8IM hPSC line enabled proliferative cell elimination within grafts. Findings demonstrate feasibility of generating a population-wide applicable, safe, off-the-shelf cell product, suitable for treating diseases for which cell-based therapies are a viable option.
Collapse
Affiliation(s)
- Chiara Pavan
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia.
| | - Kathryn C Davidson
- Infectious Diseases & Immune Defence Division, Walter and Eliza Hall Institute, Melbourne, VIC, Australia; Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Natalie Payne
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia
| | - Stefano Frausin
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
| | - Cameron P J Hunt
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
| | - Niamh Moriarty
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
| | | | - Zahra Elahi
- Department of Anatomy & Physiology, The University of Melbourne, Melbourne, VIC, Australia
| | - Andrew T Quattrocchi
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
| | | | - Le Wang
- Infectious Diseases & Immune Defence Division, Walter and Eliza Hall Institute, Melbourne, VIC, Australia; Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - William Clow
- Infectious Diseases & Immune Defence Division, Walter and Eliza Hall Institute, Melbourne, VIC, Australia; Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Huijuan Yang
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Marc Pellegrini
- Centenary Institute of Cancer Medicine and Cell Biology, Sydney, NSW, Australia
| | - Christine A Wells
- Department of Anatomy & Physiology, The University of Melbourne, Melbourne, VIC, Australia
| | - Lachlan H Thompson
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia; Faculty of Medicine and Health, School of Medical Sciences & Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Andras Nagy
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Clare L Parish
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia; Department of Anatomy & Physiology, The University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
7
|
Glamočlija S, Sabljić L, Tomić S, Đokić J, Radulović N, Gruden-Movsesijan A, Kosanović M. Trichinella spiralis extracellular vesicles induce anti-inflammatory and regulatory immune responses in vitro. Int J Parasitol 2025; 55:299-315. [PMID: 39842685 DOI: 10.1016/j.ijpara.2025.01.008] [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: 10/21/2024] [Accepted: 01/14/2025] [Indexed: 01/24/2025]
Abstract
The helminth Trichinella spiralis, through its excretory-secretory (ES L1) products, induces immune regulatory mechanisms that modulate the host's immune response not only to itself, but also to bystander antigens, foreign or self in origin, which can result in the alleviation of inflammatory diseases. Under the influence of ES L1, dendritic cells (DCs) acquire a tolerogenic phenotype and the capacity to induce Th2 and regulatory responses. Since ES L1 products represent a complex mixture of proteins and extracellular vesicles (TsEVs) the aim of this study was to investigate the impact of TsEVs, isolated from ES L1 products, on phenotypic and functional characteristics of DCs and to elucidate whether TsEVs could reproduce the immunomodulatory effects of the complete ES L1 product. Monocyte-derived DCs treated with TsEVs acquired semi-matured phenotypes, characterized by low expression of human leukocyte antigen - DR isotype (HLA-DR), cluster of differentiation (CD) 86 (CD86), and CD40, moderate expression of CD83 and C-C chemokine receptor type 7 (CCR7), and increased expression of tolerogenic markers indoleamine 2,3-dioxygenase 1 (IDO-1) and immunoglobulin-like transcript 3 (ILT3), together with the unchanged production of IL-12 and IL-23, and elevated production of IL-10 and transforming growth factor (TGF)-β, compared with controls. Gene expression analysis of TsEV-treated DCs revealed elevated levels of mTOR, Ahr, NF-κB2, RelB, SOCS1 and SOCS3, which participate in signaling pathways involved in DC maturation and the subsequent regulation of release of both anti-inflammatory and pro-inflammatory cytokines. TsEVs promoted the capacity of DCs to drive polarization of Th2 and anti-inflammatory responses, and impaired their capacity to induce Th1/Th17 polarization. Moreover, TsEV-treated DCs possessed a high capacity to induce conventional FoxP3 + regulatory T cells, as well as unconventional T regulatory (Tr1) cells. Tolerogenic properties of TsEV-treated DCs were retained even after challenge with a pro-inflammatory stimulus. These findings highlight the potential of TsEVs to induce immune tolerance, suggesting their potential use as therapeutics for the treatment of inflammatory disorders.
Collapse
Affiliation(s)
- Sofija Glamočlija
- Institute for the Application of Nuclear Energy INEP University of Belgrade Republic of Serbia
| | - Ljiljana Sabljić
- Institute for the Application of Nuclear Energy INEP University of Belgrade Republic of Serbia
| | - Sergej Tomić
- Institute for the Application of Nuclear Energy INEP University of Belgrade Republic of Serbia
| | - Jelena Đokić
- Institute of Molecular Genetics and Genetic Engineering IMGGE University of Belgrade Republic of Serbia
| | - Nataša Radulović
- Institute for Biological Research "Siniša Stanković" University of Belgrade Republic of Serbia
| | - Alisa Gruden-Movsesijan
- Institute for the Application of Nuclear Energy INEP University of Belgrade Republic of Serbia
| | - Maja Kosanović
- Institute for the Application of Nuclear Energy INEP University of Belgrade Republic of Serbia.
| |
Collapse
|
8
|
Su R, Yao T, Cao C, Yang Y, Chen M, Wu J, Zhao Y, Liu X, Li S, Ding J, Yang R, Shen S, Zhang C, Zhan C, Gao X. Enhancing Immune Responses Through Modulation of Innate Cell Microenvironments in Lymph Nodes with Virus-Mimetic Vaccines. Angew Chem Int Ed Engl 2025; 64:e202503845. [PMID: 40072248 DOI: 10.1002/anie.202503845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 03/09/2025] [Accepted: 03/12/2025] [Indexed: 03/22/2025]
Abstract
Nanovaccines hold significant promise for the prevention and treatment of infectious diseases. However, the efficacy of many nanovaccines is often limited by inadequate stimulation of both innate and adaptive immune responses. Herein, we explore a rational vaccine strategy aimed at modulating innate cell microenvironments within lymph nodes (LNs) to enhance the generation of effective immune responses. Inspired by the structure and natural infection process of viruses, we developed a versatile antigen and adjuvant co-delivery platform, termed virus-mimetic vaccines (VMVs). Specifically, polyarginine-tagged antigens were noncovalently assembled onto nucleic acid nanogels containing cytosine-phosphate-guanine oligodeoxynucleotide via a salt-bridge zipper mechanism, which can activate Toll-like receptor 9. Upon intramuscular immunization, VMVs effectively drained into the LNs, recruiting and activating multiple innate cells, including CD8+ dendritic cells (DCs), CD103+ DCs, macrophages, plasmacytoid DCs, and neutrophils. This activation modulates the innate cell microenvironments and relocates antigen-presenting cells within LNs, optimizing adaptive immune responses. VMVs induced a robust antigen-specific immune response, characterized by high levels of neutralizing antibodies, augmented memory T cell activity, and enhanced development of germinal center B cells. Together, our findings demonstrate that dynamic modulation of innate cell microenvironments by VMVs leads to optimized generation of both humoral and cellular immunity against infectious diseases.
Collapse
Affiliation(s)
- Runping Su
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Fudan University, 131 Dong An Road, Shanghai, 200032, P.R. China
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200032, P.R. China
| | - Tingting Yao
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Fudan University, 131 Dong An Road, Shanghai, 200032, P.R. China
| | - Chong Cao
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Fudan University, 131 Dong An Road, Shanghai, 200032, P.R. China
- Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P.R. China
| | - Yaqi Yang
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Fudan University, 131 Dong An Road, Shanghai, 200032, P.R. China
| | - Mingshan Chen
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Fudan University, 131 Dong An Road, Shanghai, 200032, P.R. China
| | - Jianxiao Wu
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Fudan University, 131 Dong An Road, Shanghai, 200032, P.R. China
| | - Yue Zhao
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Fudan University, 131 Dong An Road, Shanghai, 200032, P.R. China
| | - Xiaoxiao Liu
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Fudan University, 131 Dong An Road, Shanghai, 200032, P.R. China
| | - Sha Li
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Fudan University, 131 Dong An Road, Shanghai, 200032, P.R. China
| | - Junqiang Ding
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Fudan University, 131 Dong An Road, Shanghai, 200032, P.R. China
| | - Rong Yang
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Fudan University, 131 Dong An Road, Shanghai, 200032, P.R. China
- Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P.R. China
| | - Shun Shen
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Fudan University, 131 Dong An Road, Shanghai, 200032, P.R. China
| | - Chuan Zhang
- Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P.R. China
| | - Changyou Zhan
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Fudan University, 131 Dong An Road, Shanghai, 200032, P.R. China
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200032, P.R. China
| | - Xihui Gao
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Pudong Hospital, Key Laboratory of Medical Molecular Virology of MOE/NHC/CAMS, School of Basic Medical Sciences, Fudan University, 131 Dong An Road, Shanghai, 200032, P.R. China
| |
Collapse
|
9
|
Elhadary M, Elsayed B, Elshoeibi AM, Karen O, Elmakaty I, Alhmoud J, Hamdan A, Malki MI. The Clinicopathological and Prognostic Value of CCR7 Expression in Breast Cancer Throughout the Literature: A Systematic Review and Meta-Analysis. Biomedicines 2025; 13:1007. [PMID: 40299690 PMCID: PMC12024592 DOI: 10.3390/biomedicines13041007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2025] [Accepted: 04/17/2025] [Indexed: 05/01/2025] Open
Abstract
Background/Objective: This study aimed to determine the clinicopathological findings and prognostic value of chemokine receptor 7 (CCR7) expression in patients with breast cancer (BC). Methods: Up to the 25th of March 2025, a search was conducted using five databases: PubMed, Embase, Scopus, Medline, and Web of Science. The methodological standards for the epidemiological research scale were used to assess the quality of the included articles, and Stata software (Stata 19) was used to synthesize the meta-analysis. Results: We considered 12 of 853 studies that included 3119 patients with BC. High CCR7 expression was not associated with age (odds ratio [OR] 0.82, 95% confidence interval [CI] 0.66-1.03); clinicopathological findings, including tumor size (OR 1.062, 95% CI 0.630-1.791); clinical stage (OR 1.753, 95% CI 0.231-13.304); nodal metastasis (OR 1.252, 95% CI 0.571-2.741); or histological differentiation (OR 1.167, 95% CI 0.939-1.450). CCR7 expression did not affect overall survival (hazard ratio 0.996, 95% CI 0.659-1.505). Conclusions: Our quantitative analysis did not reveal an association between CCR7 expression and poor clinicopathological or prognostic features in BC patients. Because of the high heterogeneity and potential publication bias, large high-quality studies are required to further confirm these findings.
Collapse
Affiliation(s)
- Mohamed Elhadary
- College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (M.E.); (B.E.); (A.M.E.); (O.K.); (A.H.)
| | - Basel Elsayed
- College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (M.E.); (B.E.); (A.M.E.); (O.K.); (A.H.)
| | - Amgad Mohamed Elshoeibi
- College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (M.E.); (B.E.); (A.M.E.); (O.K.); (A.H.)
| | - Omar Karen
- College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (M.E.); (B.E.); (A.M.E.); (O.K.); (A.H.)
| | - Ibrahim Elmakaty
- Department of Medical Education, Hamad Medical Corporation, Doha P.O. Box 3050, Qatar;
| | - Jehad Alhmoud
- Department of Medical Laboratory Sciences, Jordan University of Science and Technology, Irbid 22110, Jordan;
| | - Ahmad Hamdan
- College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (M.E.); (B.E.); (A.M.E.); (O.K.); (A.H.)
| | - Mohammed Imad Malki
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| |
Collapse
|
10
|
Wu C, Ren Y, Li Y, Cui Y, Zhang L, Zhang P, Zhang X, Kan S, Zhang C, Xiong Y. Identification and Experimental Validation of NETosis-Mediated Abdominal Aortic Aneurysm Gene Signature Using Multi-omics, Machine Learning, and Mendelian Randomization. J Chem Inf Model 2025; 65:3771-3788. [PMID: 40105795 DOI: 10.1021/acs.jcim.4c02318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Abdominal aortic aneurysm (AAA) is a life-threatening disorder with limited therapeutic options. Neutrophil extracellular traps (NETs) are formed by a process known as "NETosis" that has been implicated in AAA pathogenesis, yet the roles and prognostic significance of NET-related genes in AAA remain poorly understood. This study aimed to identify key AAA- and NET-related genes (AAA-NETs-RGs), elucidate their potential mechanisms in contributing to AAA, and explore potential therapeutic compounds for AAA therapy. Through bioinformatics analysis of multiomics and machine learning, we identified six AAA-NETs-RGs: DUSP26, FCN1, MTHFD2, GPRC5C, SEMA4A, and CCR7, which exhibited strong diagnostic potential for predicting AAA progression, were significantly enriched in pathways related to cytokine-cytokine receptor interaction and chemokine signaling. Immune infiltration analysis revealed a causal association between AAA-NETs-RGs and immune cell infiltration. Cell-cell communication analysis indicated that AAA-NETs-RGs predominantly function in smooth muscle cells, B cells, T cells, and NK cells, primarily through cytokine and chemokine signaling. Gene profiling revealed that CCR7 and MTHFD2 exhibited the most significant upregulation in AAA patients compared to non-AAA controls, as well as in in vitro AAA models. Notably, genetic depletion of CCR7 and MTHFD2 strongly inhibited Ang II-induced phenotypic switching, functional impairment, and senescence in vascular smooth muscle cells (VSMCs). Based on AAA-NETs-RGs, molecular docking analysis combined with the Connectivity Map (CMap) database identified mirdametinib as a potential therapeutic agent for AAA. Mirdametinib effectively alleviated Ang II-induced phenotypic switching, biological dysfunction, and senescence. These findings provide valuable insights into understanding the pathophysiology of AAA and highlight promising therapeutic strategies targeting AAA-NETs-RGs.
Collapse
Affiliation(s)
- Chengsong Wu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, 710069 Xi'an, Shaanxi, P. R. China
| | - Yuanyuan Ren
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, 710069 Xi'an, Shaanxi, P. R. China
| | - Yang Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, 710069 Xi'an, Shaanxi, P. R. China
| | - Yue Cui
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, 710069 Xi'an, Shaanxi, P. R. China
| | - Liyao Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, 710069 Xi'an, Shaanxi, P. R. China
| | - Pan Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, 710069 Xi'an, Shaanxi, P. R. China
| | - Xuejiao Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, 710069 Xi'an, Shaanxi, P. R. China
| | - Shangguang Kan
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, 710069 Xi'an, Shaanxi, P. R. China
| | - Chan Zhang
- Department of Blood Transfusion, the First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, 650032 Kunming, Yunnan, China
| | - Yuyan Xiong
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, 710069 Xi'an, Shaanxi, P. R. China
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No. 3 Hospital, The Affiliated Hospital of Northwest University, 710018 Xi'an, Shaanxi, P. R. China
| |
Collapse
|
11
|
Dai X, Xi M, Li J. Cancer metastasis: molecular mechanisms and therapeutic interventions. MOLECULAR BIOMEDICINE 2025; 6:20. [PMID: 40192949 PMCID: PMC11977077 DOI: 10.1186/s43556-025-00261-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 03/07/2025] [Accepted: 03/14/2025] [Indexed: 04/10/2025] Open
Abstract
The metastatic cascade is a complicated process where cancer cells travel across multiple organs distant from their primary site of onset. Despite the wide acceptance of the 'seed and soil' theory, mechanisms driving metastasis organotropism remain mystery. Using breast cancer of different subtypes as the disease model, we characterized the 'metastatic profile of cancer cells' and the 'redox status of the organ microenvironment' as the primary determinants of cancer metastasis organotropism. Mechanically, we identified a positive correlation between cancer metabolic plasticity and stemness, and proposed oxidative stress as the selection power of cancer cells succeeding the metastasis cascade. Therapeutically, we proposed the use of pro-oxidative therapeutics in ablating cancer cells taking advantages of this fragile moment during metastasis. We comprehensively reviewed current pro-oxidative strategies for treating cancers that cover the first line chemo- and radio-therapies, approaches relying on naturally existing power including magnetic field, electric field, light and sound, nanoparticle-based anti-cancer composites obtained through artificial design, as well as cold atmospheric plasma as an innovative pro-oxidative multi-modal modality. We discussed possible combinations of pro-oxidative approaches with existing therapeutics in oncology prior to the forecast of future research directions. This paper identified the fundamental mechanics driving metastasis organotropism and proposed intervention strategies accordingly. Insights provided here may offer clues for the design of innovative solutions that may open a new paradigm for cancer treatment.
Collapse
Affiliation(s)
- Xiaofeng Dai
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China.
| | - Ming Xi
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, People's Republic of China
| | - Jitian Li
- Molecular Biology Lab, Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital), Henan Province, Zhengzhou, 450000, China
| |
Collapse
|
12
|
Lai W, Li Y, Luo OJ. MIST: An interpretable and flexible deep learning framework for single-T cell transcriptome and receptor analysis. SCIENCE ADVANCES 2025; 11:eadr7134. [PMID: 40184452 PMCID: PMC11970455 DOI: 10.1126/sciadv.adr7134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 02/28/2025] [Indexed: 04/06/2025]
Abstract
Joint analysis of transcriptomic and T cell receptor (TCR) features at single-cell resolution provides a powerful approach for in-depth T cell immune function research. Here, we introduce a deep learning framework for single-T cell transcriptome and receptor analysis, MIST (Multi-insight for T cell). MIST features three latent spaces: gene expression, TCR, and a joint latent space. Through analyses of antigen-specific T cells, and T cell datasets related to lung cancer immunotherapy and COVID19, we demonstrate MIST's interpretability and flexibility. MIST easily and accurately resolves cell function and antigen specificity by vectorizing and integrating transcriptome and TCR data of T cells. In addition, using MIST, we identified the heterogeneity of CXCL13+ subsets in lung cancer infiltrating CD8+ T cells and their association with immunotherapy, providing additional insights into the functional transition of CXCL13+ T cells related to anti-PD-1 therapy that were not reported in the original study.
Collapse
Affiliation(s)
- Wenpu Lai
- The First Affiliated Hospital, Jinan University, Guangzhou 510632, China
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou 510632, China
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Yangqiu Li
- The First Affiliated Hospital, Jinan University, Guangzhou 510632, China
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Oscar Junhong Luo
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou 510632, China
- Key Laboratory of Viral Pathogenesis and Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China
- Zhuhai Institute of Jinan University, Jinan University, Zhuhai 519070, China
| |
Collapse
|
13
|
He X, Huang J, Ma H, Ma Z, Huang C, Ling Y, Zhou B, Li J. Identification of novel IL17-related genes as prognostic and therapeutic biomarkers of psoriasis using comprehensive bioinformatics analysis and machine learning. Sci Rep 2025; 15:11295. [PMID: 40175394 PMCID: PMC11965382 DOI: 10.1038/s41598-025-87556-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 01/20/2025] [Indexed: 04/04/2025] Open
Abstract
Psoriasis is a common chronic skin disorder with a polygenic background. It is widely acknowledged that Th17/IL-17A axis plays a key role in the pathogenesis of psoriasis. However, numerous regulatory genes upstream of the pathway remain undiscovered, creating a knowledge gap in our understanding of the genetic aspects of the Th17/IL-17A axis. In this study, we employed machine learning algorithms to identify three target genes associated with psoriasis: CCR7, IL2RG, and PLEK. The validation of these genes was carried out in specimens from psoriatic patients. In vivo, investigations assessed the relationship between these three genes and IL-17A-related inflammation and their connection to psoriatic phenotypes. To further confirm the significance of the newly discovered gene, PLEK, we performed experiments involving the blockade of its expression. Our bioinformatics analysis revealed three novel genes closely linked to psoriasis: CCR7, IL2RG, and PLEK. These genes exhibited upregulated expression in psoriasis, consistently aligning with the Th17/IL-17A axis. Inhibition of PLEK expression reduced Th17/IL-17A-related inflammation and alleviated psoriatic phenotypes. CCR7, IL2RG, and PLEK show potential as three novel biomarkers for psoriasis, with PLEK being reported for the first time in this context. These genes contribute to pathogenesis by associating with the Th17/IL-17A signaling pathway.
Collapse
Affiliation(s)
- Xingling He
- Yiling Hospital of Yichang City, Yichang, 443000, Hubei, China
| | - Jingjing Huang
- Yiling Hospital of Yichang City, Yichang, 443000, Hubei, China
| | - Hanying Ma
- School of Life Sciences, Huanggang Normal University, Huanggang, 438000, China
- College of Biology Pharmacy, Three Gorges University, Yichang, 443000, Hubei, China
| | - Zhujun Ma
- College of Biology Pharmacy, Three Gorges University, Yichang, 443000, Hubei, China
| | - Changzheng Huang
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Yunli Ling
- Beijing Huairou Hospital, Capital Medical University, Beijing, 101400, China.
| | - Bin Zhou
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China.
- Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Jingang Li
- Yichang Central People's Hospital, the first College of Clinical Medical Science, Three Gorges University, Yichang, 443000, Hubei, China.
| |
Collapse
|
14
|
Eskandari A, Leow TC, Rahman MBA, Oslan SN. Advances in Therapeutic Cancer Vaccines, Their Obstacles, and Prospects Toward Tumor Immunotherapy. Mol Biotechnol 2025; 67:1336-1366. [PMID: 38625508 DOI: 10.1007/s12033-024-01144-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 03/15/2024] [Indexed: 04/17/2024]
Abstract
Over the past few decades, cancer immunotherapy has experienced a significant revolution due to the advancements in immune checkpoint inhibitors (ICIs) and adoptive cell therapies (ACTs), along with their regulatory approvals. In recent times, there has been hope in the effectiveness of cancer vaccines for therapy as they have been able to stimulate de novo T-cell reactions against tumor antigens. These tumor antigens include both tumor-associated antigen (TAA) and tumor-specific antigen (TSA). Nevertheless, the constant quest to fully achieve these abilities persists. Therefore, this review offers a broad perspective on the existing status of cancer immunizations. Cancer vaccine design has been revolutionized due to the advancements made in antigen selection, the development of antigen delivery systems, and a deeper understanding of the strategic intricacies involved in effective antigen presentation. In addition, this review addresses the present condition of clinical tests and deliberates on their approaches, with a particular emphasis on the immunogenicity specific to tumors and the evaluation of effectiveness against tumors. Nevertheless, the ongoing clinical endeavors to create cancer vaccines have failed to produce remarkable clinical results as a result of substantial obstacles, such as the suppression of the tumor immune microenvironment, the identification of suitable candidates, the assessment of immune responses, and the acceleration of vaccine production. Hence, there are possibilities for the industry to overcome challenges and enhance patient results in the coming years. This can be achieved by recognizing the intricate nature of clinical issues and continuously working toward surpassing existing limitations.
Collapse
Affiliation(s)
- Azadeh Eskandari
- Enzyme and Microbial Technology Research Centre, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia.
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia.
| | - Thean Chor Leow
- Enzyme and Microbial Technology Research Centre, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
- Enzyme Technology and X-ray Crystallography Laboratory, VacBio 5, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
| | | | - Siti Nurbaya Oslan
- Enzyme and Microbial Technology Research Centre, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
- Enzyme Technology and X-ray Crystallography Laboratory, VacBio 5, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
| |
Collapse
|
15
|
Zhao Y, Tian M, Tong X, Yang X, Gan L, Yong T. Emerging strategies in lymph node-targeted nano-delivery systems for tumor immunotherapy. Essays Biochem 2025; 69:EBC20253008. [PMID: 40159756 DOI: 10.1042/ebc20253008] [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: 01/13/2025] [Accepted: 02/26/2025] [Indexed: 04/02/2025]
Abstract
The emergence of immunotherapy has led to the clinical approval of several related drugs. However, their efficacy against solid tumors remains limited. As the hub of immune activation, lymph nodes (LNs) play a critical role in tumor immunotherapy by initiating and amplifying immune responses. Nevertheless, the intricate physiological structure and barriers within LNs, combined with the immunosuppressive microenvironment induced by tumor cells, significantly impede the therapeutic efficacy of immunotherapy. Engineered nanoparticles (NPs) have shown great potential in overcoming these challenges by facilitating targeted drug transport to LNs and directly or indirectly activating T cells. This review systematically examines the structural features of LNs, key factors influencing the targeting efficiency of NPs, and current strategies for remodeling the immunosuppressive microenvironment of LNs. Additionally, it discusses future opportunities for optimizing NPs to enhance tumor immunotherapy, addressing challenges in clinical translation and safety evaluation.
Collapse
Affiliation(s)
- Yaoli Zhao
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Muzi Tian
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xin Tong
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Lu Gan
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Tuying Yong
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
16
|
Ando H, Tsubouchi K, Yanagihara T, Hata K, Eto D, Suzuki K, Hamada N, Okamoto I. Involvement of naïve T cells in the pathogenesis of osimertinib-induced pneumonitis. Sci Rep 2025; 15:10545. [PMID: 40148432 PMCID: PMC11950390 DOI: 10.1038/s41598-025-95271-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 03/20/2025] [Indexed: 03/29/2025] Open
Abstract
Osimertinib, a third-generation epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor, effectively treats EGFR-mutated non-small cell lung cancer. Drug-induced interstitial lung disease, a potentially fatal complication of osimertinib, involves an increase in lymphocytes in the bronchoalveolar lavage fluid (BALF). As the precise role of these lymphocytes is unknown, we investigated the pathogenesis of osimertinib-induced pneumonitis using BALF obtained from patients, and an osimertinib-induced pneumonitis mouse model. Mass cytometry revealed the presence of CCR7+ CD45RA+ naïve T cells in the BALF of patients with osimertinib-induced pneumonitis. Body weight measurements, BALF analysis, histopathological evaluation and RNA sequencing of mouse lung tissue were performed to investigate immune cell involvement and the mechanisms underlying osimertinib-induced pneumonitis. In the mouse model, administration of osimertinib after naphthalene-induced damage to the bronchiolar epithelium exacerbated lung inflammation and resulted in significant weight loss. Immunofluorescence staining revealed the infiltration of CCR7+ cells into the lungs of mice treated with naphthalene and osimertinib. Bulk RNA sequencing identified an upregulation of chemokine-related biological processes, with increased expression of C-C motif chemokine ligand 21 (Ccl21) and C-C motif chemokine ligand 8 (Ccl8) in the lung tissue. Additionally, immunofluorescence staining confirmed elevated expression of Ccl21 and Ccl8 in the distal bronchiolar epithelium. This study provides insights into the mechanisms underlying osimertinib-induced pneumonitis.
Collapse
Affiliation(s)
- Hiroyuki Ando
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Kazuya Tsubouchi
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan.
| | - Toyoshi Yanagihara
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Kentaro Hata
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Daisuke Eto
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Kunihiro Suzuki
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Naoki Hamada
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
- Department of Respiratory Medicine, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Isamu Okamoto
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| |
Collapse
|
17
|
Li J, Yang R, Zhang C, Lovell JF, Zhang Y. Manganese-Driven Plasmid Nanofibers Formed In Situ for Cancer Gene Delivery and Metalloimmunotherapy. J Am Chem Soc 2025; 147:10504-10518. [PMID: 40065259 DOI: 10.1021/jacs.4c18511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
While nucleic-acid-based cancer vaccines hold therapeutic potential, their limited immunogenicity remains a challenge due in part to the low efficiency of cytoplasmic delivery caused by lysosomal entrapment. In this work, we found that plasmids encoding both an antigen and a STING agonist protein adjuvant can self-assemble into coordination nanofibers, triggered by manganese ions. We developed a strategy to construct a DNA vaccine, termed MnO2-OVA-CDA-mem, formed by the coencapsulation of manganese dioxide (MnO2), an antigen-expressing plasmid (encoding ovalbumin, OVA), and an adjuvant enzyme-expressing plasmid (encoding STING agonist, CDA) within dendritic cell (DC) membranes. Upon uptake into acidic lysosomes, Mn2+ released from MnO2 triggered the nucleic acids to undergo a morphological change from nanospheres (∼180 nm diameter) to nanofibers (∼1 μm length), resulting in an increase in mechanical strength by about 9-fold and consequently lysosomal membrane disruption. The antigen OVA and adjuvants Mn2+ and CDA in the cytoplasm triggered strong DC activation and antigen-specific CD8+ T cell metalloimmune responses, significantly inhibiting the growth of B16-OVA tumors and inducing long-term immune memory. Altogether, MnO2-OVA-CDA-mem holds potential as a platform for nucleic acid antigen and adjuvant delivery using an in situ self-assembly strategy in a metal-driven, stimulus-responsive, and programmable manner for cancer metalloimmunotherapy.
Collapse
Affiliation(s)
- Jiexin Li
- School of Chemical Engineering and Technology, State Key Laboratory of Synthetic Biology, Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, P. R. China, 300350
| | - Ruiqi Yang
- School of Chemical Engineering and Technology, State Key Laboratory of Synthetic Biology, Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, P. R. China, 300350
| | - Chen Zhang
- School of Chemical Engineering and Technology, State Key Laboratory of Synthetic Biology, Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, P. R. China, 300350
| | - Jonathan F Lovell
- Department of Biomedical Engineering, The State University of New York at Buffalo, Buffalo, New York 14260, United States
| | - Yumiao Zhang
- School of Chemical Engineering and Technology, State Key Laboratory of Synthetic Biology, Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, P. R. China, 300350
| |
Collapse
|
18
|
Sun Y, Qian Y, Qiu L, Zhu X, Ning H, Pang L, Niu X, Liu Y, Zhou X, Chen G, Zhai W, Gao Y. A novel peptide targeting CCR7 inhibits tumor cell lymph node metastasis. Cancer Immunol Immunother 2025; 74:153. [PMID: 40105966 PMCID: PMC11923353 DOI: 10.1007/s00262-025-03995-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 02/21/2025] [Indexed: 03/22/2025]
Abstract
Lymph nodes are the most common metastasis sites for tumor cells, which are intimately linked to patient prognosis. It has been reported that cancer cells can upregulate CC Chemokine Receptor 7 (CCR7) expression and hijack its normal functions, enabling them to migrate along the gradient of CCL19 and CCL21 toward the lymph nodes and colonies as the initial stage of distant metastasis. In tumor patients, the metastatic tumor in the lymph nodes exhibited higher expression of CCR7, as well as inhibitory immune checkpoints PD-1, LAG-3, and TIM-3 compared to the primary tumors with the analysis of TCGA and GEO databases. Also, in mouse tumor model, tumor cells with elevated CCR7 expression were more susceptible to develop popliteal lymph node metastasis. Subsequently, we successfully identified a CCR7 binding peptide TC6 by phage display biopanning, which specifically blocks the interaction of CCR7/CCL19 and CCR7/CCL21. Further, the D-amino acids were introduced to substitute the N- and C-terminus of TC6 peptide to obtain the proteolysis-resistant TC6-D3 peptide, which decreased tumor cell migration in vitro via ERK1/2 pathway and inhibited tumor growth and lymph nodes metastasis in vivo, as well as effectively restored T cells cytotoxicity in both primary tumors and lymph nodes. In conclusion, CCR7 promoted tumor cell metastasis to lymph node and inhibited the anti-tumor immune responses in lymph nodes. Specific blockade of the CCR7 pathway with TC6-D3 peptide can significantly reduce lymph node tumor burden, promoting CD8+ T cell infiltration in primary tumors, meanwhile, enhancing anti-tumor immune responses in lymph nodes.
Collapse
Affiliation(s)
- Yixuan Sun
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Yuzhen Qian
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Lu Qiu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Xueqin Zhu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Haoming Ning
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Liwei Pang
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Xiaoshuang Niu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Yi Liu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Xiuman Zhou
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Guanyu Chen
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Wenjie Zhai
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| | - Yanfeng Gao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| |
Collapse
|
19
|
Gao M, Wang X, Su S, Feng W, Lai Y, Huang K, Cao D, Wang Q. Meningeal lymphatic vessel crosstalk with central nervous system immune cells in aging and neurodegenerative diseases. Neural Regen Res 2025; 20:763-778. [PMID: 38886941 PMCID: PMC11433890 DOI: 10.4103/nrr.nrr-d-23-01595] [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: 09/21/2023] [Revised: 10/30/2023] [Accepted: 12/22/2023] [Indexed: 06/20/2024] Open
Abstract
Meningeal lymphatic vessels form a relationship between the nervous system and periphery, which is relevant in both health and disease. Meningeal lymphatic vessels not only play a key role in the drainage of brain metabolites but also contribute to antigen delivery and immune cell activation. The advent of novel genomic technologies has enabled rapid progress in the characterization of myeloid and lymphoid cells and their interactions with meningeal lymphatic vessels within the central nervous system. In this review, we provide an overview of the multifaceted roles of meningeal lymphatic vessels within the context of the central nervous system immune network, highlighting recent discoveries on the immunological niche provided by meningeal lymphatic vessels. Furthermore, we delve into the mechanisms of crosstalk between meningeal lymphatic vessels and immune cells in the central nervous system under both homeostatic conditions and neurodegenerative diseases, discussing how these interactions shape the pathological outcomes. Regulation of meningeal lymphatic vessel function and structure can influence lymphatic drainage, cerebrospinal fluid-borne immune modulators, and immune cell populations in aging and neurodegenerative disorders, thereby playing a key role in shaping meningeal and brain parenchyma immunity.
Collapse
Affiliation(s)
- Minghuang Gao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Xinyue Wang
- The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Shijie Su
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Weicheng Feng
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Yaona Lai
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Kongli Huang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Dandan Cao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Qi Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| |
Collapse
|
20
|
Lu X, Lin W, Zheng J, Huang W, Yu S, Chen H, Wang H, Zhang Y. Sodium nitrite orchestrates macrophage mimicry of tongue squamous carcinoma cells to drive lymphatic metastasis. Br J Cancer 2025; 132:340-353. [PMID: 39799274 PMCID: PMC11833070 DOI: 10.1038/s41416-024-02923-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 11/21/2024] [Accepted: 11/28/2024] [Indexed: 01/15/2025] Open
Abstract
BACKGROUND Tongue squamous cell carcinoma (TSCC) is a malignant oral cancer with unclear pathogenesis that shows a tendency for early-stage lymphatic metastasis. This results in a poor prognosis, with a low 5-year survival rate. Dietary sodium nitrite (NaNO2) has proposed associations with disease, including cancer. However, a direct relationship between NaNO2 and TSCC has not been established. METHODS In vitro and in vivo assays were used to investigate the role of NaNO2 in TSCC. Protein expression in TSCC specimens was detected by immunohistochemistry and immunofluorescence. The molecular mechanism was determined using RT-qPCR, western blot, RNA-seq, luciferase reporter assays, migration assays, and FACS analysis. More detail of methods can be found in the Materials and methods section. RESULTS The data in this study showed that NaNO2 did not initiate carcinogenesis in the tongue but improved the lymphatic metastatic potential of TSCC cells in the specified experimental period. During metastasis to lymph nodes, monocyte-macrophage markers were upregulated in TSCC cells, whereas keratin markers were downregulated. Specifically, expression of the CD68 gene was high in TSCC cells following NaNO2-induced TSCC phenotypic switching. These phenotypic changes were associated with activation of transcription factor cyclic-AMP response binding protein (CREB1), which directly targets CD68 transcription. Furthermore, blocking CREB1 activity either through gene knockout or specific inhibitor treatment decreased the migration ability of TSCC cells and suppressed CD68 expression. CONCLUSIONS Our findings highlight the role of NaNO2 in enabling macrophage mimicry in TSCC cells through the CREB1-CD68 signaling pathway, which promotes lymphatic metastasis. Shedding light on drivers of lymphatic metastasis in TSCC and providing a new perspective on dietary strategies to improve outcomes of patients with TSCC.
Collapse
Affiliation(s)
- Xiangwan Lu
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, PR China
| | - Weifan Lin
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, PR China
| | - Junheng Zheng
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, PR China
| | - Wuheng Huang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, PR China
| | - Shuyi Yu
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, PR China
| | - Haoxiang Chen
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, PR China
| | - Hua Wang
- Department of Oral and Maxillofacial Surgery, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, PR China.
| | - Yan Zhang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, PR China.
| |
Collapse
|
21
|
Satofuka H, Suzuki H, Tanaka T, Ubukata R, Hirose M, Yamamoto H, Kaneko Y, Fujisawa S, Li G, Kaneko MK, Kato Y. A novel anti-mouse CCR7 monoclonal antibody, C 7Mab-7, demonstrates high sensitivity in flow cytometry, western blot, and immunohistochemistry. Biochem Biophys Rep 2025; 41:101948. [PMID: 40028039 PMCID: PMC11870228 DOI: 10.1016/j.bbrep.2025.101948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/10/2025] [Accepted: 02/05/2025] [Indexed: 03/05/2025] Open
Abstract
C-C chemokine receptor type 7 (CCR7) is a member of the G protein-coupled receptor family and functions as a lymph node-homing receptor for immune cells. Upon ligand binding, CCR7 promotes the migration of immune cells to secondary lymphoid organs. In cancers, CCR7 has been revealed as a critical molecule in lymph node metastasis. Consequently, anti-CCR7 monoclonal antibodies (mAbs) have been developed as cancer therapeutic agents. In this study, we established an anti-mouse CCR7 (mCCR7) mAb, C7Mab-7 (rat IgG1, kappa) using the Cell-Based Immunization and Screening (CBIS) method. C7Mab-7 demonstrated high sensitivity in flow cytometry. The dissociation constant (K D) value of C7Mab-7 was determined to be 2.5 × 10⁻⁹ M for mCCR7-overexpressed Chinese hamster ovary-K1 (CHO/mCCR7) cells. Furthermore, C7Mab-7 detected mCCR7 with high sensitivity in western blot and immunohistochemistry. C7Mab-7, developed by the CBIS method, accelerates the development of CCR7-targeted antibody therapies and cancer diagnostics.
Collapse
Affiliation(s)
- Hiroyuki Satofuka
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Hiroyuki Suzuki
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Tomohiro Tanaka
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Rena Ubukata
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Miu Hirose
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Haruto Yamamoto
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Yu Kaneko
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Shiori Fujisawa
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Guanjie Li
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Mika K. Kaneko
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Yukinari Kato
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| |
Collapse
|
22
|
Meloun A, León B. Beyond CCR7: dendritic cell migration in type 2 inflammation. Front Immunol 2025; 16:1558228. [PMID: 40093008 PMCID: PMC11906670 DOI: 10.3389/fimmu.2025.1558228] [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: 01/09/2025] [Accepted: 02/13/2025] [Indexed: 03/19/2025] Open
Abstract
Conventional dendritic cells (cDCs) are crucial antigen-presenting cells that initiate and regulate T cell responses, thereby shaping immunity against pathogens, innocuous antigens, tumors, and self-antigens. The migration of cDCs from peripheral tissues to draining lymph nodes (dLNs) is essential for their function in immune surveillance. This migration allows cDCs to convey the conditions of peripheral tissues to antigen-specific T cells in the dLNs, facilitating effective immune responses. Migration is primarily mediated by chemokine receptor CCR7, which is upregulated in response to homeostatic and inflammatory cues, guiding cDCs to dLNs. However, during type 2 immune responses, such as those triggered by parasites or allergens, a paradox arises-cDCs exhibit robust migration to dLNs despite low CCR7 expression. This review discusses how type 2 inflammation relies on additional signaling pathways, including those induced by membrane-derived bioactive lipid mediators like eicosanoids, sphingolipids, and oxysterols, which cooperate with CCR7 to enhance cDC migration and T helper 2 (Th2) differentiation. We explore the potential regulatory mechanisms of cDC migration in type 2 immunity, offering insights into the differential control of cDC trafficking in diverse immune contexts and its impact on immune responses.
Collapse
Affiliation(s)
- Audrey Meloun
- Innate Cells and Th2 Immunity Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Beatriz León
- Innate Cells and Th2 Immunity Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
23
|
Wurnig S, Huber ME, Weiler C, Baltrukevich H, Merten N, Stötzel I, Steffen T, Chang Y, Klammer RH, Baumjohann D, Kiermaier E, Kolb P, Kostenis E, Schiedel M, Hansen FK. A Fluorescent Probe Enables the Discovery of Improved Antagonists Targeting the Intracellular Allosteric Site of the Chemokine Receptor CCR7. J Med Chem 2025; 68:4308-4333. [PMID: 39937529 PMCID: PMC11873976 DOI: 10.1021/acs.jmedchem.4c02102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 01/04/2025] [Accepted: 01/27/2025] [Indexed: 02/13/2025]
Abstract
Intracellular ligands of G protein-coupled receptors (GPCRs) are gaining significant interest in drug discovery. Here, we report the development of the fluorescent ligand Mz437 (4) targeting the CC chemokine receptor CCR7 at an intracellular allosteric site. We demonstrate its experimental power by applying 4 to identify two improved intracellular CCR7 antagonists, SLW131 (10) and SLW132 (21m), developed by converting two weakly active antagonists into single- or double-digit nanomolar ligands with minimal modifications. The thiadiazoledioxide 10 was derived from the CCR7 antagonist Cmp2105 by removing a methyl group from the benzamide moiety, while the squaramide 21m was obtained from the CXCR1/CXCR2 antagonist and clinical candidate navarixin by replacing the ethyl substituent by a tert-butyl group to engage a lipophilic subpocket. We show that 10 and 21m qualify to probe CCR7 biology in recombinant and primary immune cells and expect our novel probes to facilitate the design of next-generation intracellular CCR7 ligands.
Collapse
Affiliation(s)
- Silas
L. Wurnig
- Department
of Pharmaceutical & Cell Biological Chemistry, Pharmaceutical
Institute, University of Bonn, An der Immenburg 4, Bonn 53121, Germany
| | - Max E. Huber
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-University Erlangen-Nürnberg, Nikolaus-Fiebiger-Straße 10, Erlangen 91058, Germany
| | - Corinna Weiler
- Molecular,
Cellular and Pharmacobiology Section, Institute for Pharmaceutical
Biology, University of Bonn, Nussallee 6, Bonn 53115, Germany
| | - Hanna Baltrukevich
- Department
of Pharmaceutical Chemistry, University
of Marburg, Marbacher Weg 8, Marburg 35037, Germany
| | - Nicole Merten
- Molecular,
Cellular and Pharmacobiology Section, Institute for Pharmaceutical
Biology, University of Bonn, Nussallee 6, Bonn 53115, Germany
| | - Isabel Stötzel
- Life
and Medical Sciences (LIMES) Institute, Immune and Tumor Biology, University of Bonn, Bonn 53115, Germany
| | - Teresa Steffen
- Medical
Clinic III for Oncology, Hematology, Immuno-Oncology and Rheumatology,
University Hospital Bonn, University of
Bonn, Venusberg-Campus
1, Bonn 53127, Germany
| | - Yinshui Chang
- Medical
Clinic III for Oncology, Hematology, Immuno-Oncology and Rheumatology,
University Hospital Bonn, University of
Bonn, Venusberg-Campus
1, Bonn 53127, Germany
| | - René H.
L. Klammer
- Department
of Pharmaceutical & Cell Biological Chemistry, Pharmaceutical
Institute, University of Bonn, An der Immenburg 4, Bonn 53121, Germany
| | - Dirk Baumjohann
- Medical
Clinic III for Oncology, Hematology, Immuno-Oncology and Rheumatology,
University Hospital Bonn, University of
Bonn, Venusberg-Campus
1, Bonn 53127, Germany
| | - Eva Kiermaier
- Life
and Medical Sciences (LIMES) Institute, Immune and Tumor Biology, University of Bonn, Bonn 53115, Germany
| | - Peter Kolb
- Department
of Pharmaceutical Chemistry, University
of Marburg, Marbacher Weg 8, Marburg 35037, Germany
| | - Evi Kostenis
- Molecular,
Cellular and Pharmacobiology Section, Institute for Pharmaceutical
Biology, University of Bonn, Nussallee 6, Bonn 53115, Germany
| | - Matthias Schiedel
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-University Erlangen-Nürnberg, Nikolaus-Fiebiger-Straße 10, Erlangen 91058, Germany
- Institute
of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig, Beethovenstraße 55, Braunschweig 38106, Germany
| | - Finn K. Hansen
- Department
of Pharmaceutical & Cell Biological Chemistry, Pharmaceutical
Institute, University of Bonn, An der Immenburg 4, Bonn 53121, Germany
| |
Collapse
|
24
|
Hahn H, Daly C, Little J, Perry-Hauser NA, Flores-Espinoza E, Inoue A, Plouffe B, Thomsen ARB. Endosomal chemokine receptor signalosomes regulate central mechanisms underlying cell migration. eLife 2025; 13:RP99373. [PMID: 39992711 PMCID: PMC11850004 DOI: 10.7554/elife.99373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2025] Open
Abstract
Chemokine receptors are GPCRs that regulate the chemotactic migration of a wide variety of cells including immune and cancer cells. Most chemokine receptors contain features associated with the ability to stimulate G protein signaling during β-arrestin-mediated receptor internalization into endosomes. As endosomal signaling of certain non-GPCR receptors plays a major role in cell migration, we chose to investigate the potential role of endosomal chemokine receptor signaling on mechanisms governing this function. Applying a combination of pharmacological and cell biological approaches, we demonstrate that the model chemokine receptor CCR7 recruits G protein and β-arrestin simultaneously upon chemokine stimulation, which enables internalized receptors to activate G protein from endosomes. Furthermore, spatiotemporal-resolved APEX2 proteome profiling shows that endosomal CCR7 uniquely enriches specific Rho GTPase regulators as compared to plasma membrane CCR7, which is directly associated with enhanced activity of the Rho GTPase Rac1 and chemotaxis of immune T cells. As Rac1 drives the formation of membrane protrusions during chemotaxis, our findings suggest an important integrated function of endosomal chemokine receptor signaling in cell migration.
Collapse
Affiliation(s)
- Hyunggu Hahn
- Department of Molecular Pathobiology, New York University College of DentistryNew YorkUnited States
- NYU Pain Research Center, New York University College of DentistryNew YorkUnited States
| | - Carole Daly
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University BelfastBelfastUnited Kingdom
| | - John Little
- Department of Molecular Pathobiology, New York University College of DentistryNew YorkUnited States
- Department of Surgery, Columbia University Columbia University Vagelos College of Physicians and SurgeonsNew YorkUnited States
| | - Nicole A Perry-Hauser
- Department of Surgery, Columbia University Columbia University Vagelos College of Physicians and SurgeonsNew YorkUnited States
| | - Emmanuel Flores-Espinoza
- Department of Molecular Pathobiology, New York University College of DentistryNew YorkUnited States
- NYU Pain Research Center, New York University College of DentistryNew YorkUnited States
| | - Asuka Inoue
- Graduate School of Pharmaceutical Science, Tohoku UniversitySendaiJapan
- Graduate School of Pharmaceutical Science, Kyoto UniversityKyotoJapan
| | - Bianca Plouffe
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University BelfastBelfastUnited Kingdom
| | - Alex RB Thomsen
- Department of Molecular Pathobiology, New York University College of DentistryNew YorkUnited States
- NYU Pain Research Center, New York University College of DentistryNew YorkUnited States
- Department of Surgery, Columbia University Columbia University Vagelos College of Physicians and SurgeonsNew YorkUnited States
| |
Collapse
|
25
|
Chen KY, De Giovanni M, Xu Y, An J, Kirthivasan N, Lu E, Jiang K, Brooks S, Ranucci S, Yang J, Kanameishi S, Kabashima K, Brulois K, Bscheider M, Butcher EC, Cyster JG. Inflammation switches the chemoattractant requirements for naive lymphocyte entry into lymph nodes. Cell 2025; 188:1019-1035.e22. [PMID: 39708807 PMCID: PMC11845304 DOI: 10.1016/j.cell.2024.11.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 08/27/2024] [Accepted: 11/19/2024] [Indexed: 12/23/2024]
Abstract
Sustained lymphocyte migration from blood into lymph nodes (LNs) is important for immune responses. The CC-chemokine receptor-7 (CCR7) ligand CCL21 is required for LN entry but is downregulated during inflammation, and it has been unclear how recruitment is maintained. Here, we show that the oxysterol biosynthetic enzyme cholesterol-25-hydroxylase (Ch25h) is upregulated in LN high endothelial venules during viral infection. Lymphocytes become dependent on oxysterols, generated through a transcellular endothelial-fibroblast metabolic pathway, and the receptor EBI2 for inflamed LN entry. Additionally, Langerhans cells are an oxysterol source. Ch25h is also expressed in inflamed peripheral endothelium, and EBI2 mediates B cell recruitment in a tumor model. Finally, we demonstrate that LN CCL19 is critical in lymphocyte recruitment during inflammation. Thus, our work explains how naive precursor trafficking is sustained in responding LNs, identifies a role for oxysterols in cell recruitment into inflamed tissues, and establishes a logic for the CCR7 two-ligand system.
Collapse
Affiliation(s)
- Kevin Y Chen
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA; Medical Scientist Training Program, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Marco De Giovanni
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ying Xu
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jinping An
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Nikhita Kirthivasan
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Erick Lu
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kan Jiang
- Biodata Mining and Discovery Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Stephen Brooks
- Biodata Mining and Discovery Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Serena Ranucci
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jiuling Yang
- Department of Urology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Shuto Kanameishi
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Kenji Kabashima
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Kevin Brulois
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
| | - Michael Bscheider
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
| | - Eugene C Butcher
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
| | - Jason G Cyster
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
26
|
Brisse ME, Hickman HD. Viral Infection and Dissemination Through the Lymphatic System. Microorganisms 2025; 13:443. [PMID: 40005808 PMCID: PMC11858409 DOI: 10.3390/microorganisms13020443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 02/11/2025] [Accepted: 02/15/2025] [Indexed: 02/27/2025] Open
Abstract
Many viruses induce viremia (virus in the blood) and disseminate throughout the body via the bloodstream from the initial infection site. However, viruses must often pass through the lymphatic system to reach the blood. The lymphatic system comprises a network of vessels distinct from blood vessels, along with interconnected lymph nodes (LNs). The complex network has become increasingly appreciated as a crucial host factor that contributes to both the spread and control of viral infections. Viruses can enter the lymphatics as free virions or along with migratory cells. Once virions arrive in the LN, sinus-resident macrophages remove infectious virus from the lymph. Depending on the virus, macrophages can eliminate infection or propagate the virus. A virus released from an LN is eventually deposited into the blood. This unique pathway highlights LNs as targets for viral infection control and for modulation of antiviral response development. Here, we review the lymphatic system and viruses that disseminate through this network. We discuss infection of the LN, the generation of adaptive antiviral immunity, and current knowledge of protection within the infected node. We conclude by sharing insights from ongoing efforts to optimize lymphatic targeting by vaccines and pharmaceuticals. Understanding the lymphatic system's role during viral infection enhances our knowledge of antiviral immunity and virus-host interactions and reveals potential targets for next-generation therapies.
Collapse
Affiliation(s)
| | - Heather D. Hickman
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20852, USA;
| |
Collapse
|
27
|
Morel E, Herranz-Jusdado JG, Simón R, Vicente-Gil S, González L, Tafalla C. CCR7A defines a subpopulation of IgD +IgM - B cells with higher IgD secreting capacity in the rainbow trout skin. Front Immunol 2025; 16:1538234. [PMID: 40028335 PMCID: PMC11868095 DOI: 10.3389/fimmu.2025.1538234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 01/28/2025] [Indexed: 03/05/2025] Open
Abstract
B cells exclusively expressing IgD on the cell surface (IgD+IgM- B cells) have been identified in mammals, where they seem to play a still not well-defined role in peripheral tolerance. These cells have also been reported in catfish (Ictalurus punctatus) peripheral blood and in several mucosal tissues of rainbow trout (Oncorhynchus mykiss), including gut, gills and skin. As in mammals, the precise function of these cells remains obscure, yet, in rainbow trout mucosal surfaces, these cells have been shown to be differentiated to plasma-like cells. Interestingly, in the gills, these IgD+IgM- B cells expressed high levels of the CC chemokine receptor 7 (CCR7), receptor that in mammals controls the migration of B and T cells to secondary lymphoid organs. In this work, we have established that this is also true for the trout skin, where CCR7 defines a specific subset of IgD+IgM- B cells that are further differentiated to a plasma-like profile than those not expressing CCR7. These findings increase the current understanding of this enigmatic B cell population and point to CCR7 as a key differentiation marker for these cells.
Collapse
Affiliation(s)
- Esther Morel
- Animal Biotechnology Department, National Institute of Agricultural and Food Research and Technology (INIA), Spanish Research Council (CSIC), Madrid, Spain
- Animal Health Research Center (CISA), National Institute of Agricultural and Food Research and Technology (INIA), Spanish Research Council (CSIC), Madrid, Spain
| | - Juan German Herranz-Jusdado
- Animal Health Research Center (CISA), National Institute of Agricultural and Food Research and Technology (INIA), Spanish Research Council (CSIC), Madrid, Spain
- Skretting Aquaculture Innovation, Stavanger, Norway
| | - Rocío Simón
- Animal Health Research Center (CISA), National Institute of Agricultural and Food Research and Technology (INIA), Spanish Research Council (CSIC), Madrid, Spain
| | - Samuel Vicente-Gil
- Animal Biotechnology Department, National Institute of Agricultural and Food Research and Technology (INIA), Spanish Research Council (CSIC), Madrid, Spain
| | - Lucía González
- Animal Biotechnology Department, National Institute of Agricultural and Food Research and Technology (INIA), Spanish Research Council (CSIC), Madrid, Spain
- Animal Health Research Center (CISA), National Institute of Agricultural and Food Research and Technology (INIA), Spanish Research Council (CSIC), Madrid, Spain
| | - Carolina Tafalla
- Animal Biotechnology Department, National Institute of Agricultural and Food Research and Technology (INIA), Spanish Research Council (CSIC), Madrid, Spain
| |
Collapse
|
28
|
Chen S, Wang H, Zhang L, Xi Y, Lu Y, Yu K, Zhu Y, Regina I, Bi Y, Tong F. Glymphatic system: a self-purification circulation in brain. Front Cell Neurosci 2025; 19:1528995. [PMID: 40012567 PMCID: PMC11861344 DOI: 10.3389/fncel.2025.1528995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 01/30/2025] [Indexed: 02/28/2025] Open
Abstract
The glymphatic system theory introduces a new perspective on fluid flow and homeostasis in the brain. Here, cerebrospinal fluid and interstitial fluid (CSF-ISF) moves from the perivascular spaces (PVS) of arteries to those of veins for drainage. Aquaporin-4 (AQP4) plays a crucial role in driving fluid within the PVS. The impairment to AQP4 is closely linked to the dysfunction of the glymphatic system. The function of the glymphatic system is less active during waking but enhanced during sleep. The efficiency of the glymphatic system decreases with aging. Damage to the glymphatic system will give rise to the development and progression of many brain diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), chronic traumatic encephalopathy (CTE), and vascular dementia (VaD). Here, we reviewed previous research associated with the glymphatic system, including its concepts, principles, and influencing factors. We hypothesize that AQP4 could be a target for the prevention and treatment of certain brain diseases through the regulation on the glymphatic system.
Collapse
Affiliation(s)
- Siying Chen
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
- Institute of Wound Prevention and Treatment, School of Fundamental Medicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Huijing Wang
- Institute of Wound Prevention and Treatment, School of Fundamental Medicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Lini Zhang
- Institute of Wound Prevention and Treatment, School of Fundamental Medicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Yingying Xi
- Institute of Wound Prevention and Treatment, School of Fundamental Medicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Yiying Lu
- Institute of Wound Prevention and Treatment, School of Fundamental Medicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Kailin Yu
- Institute of Wound Prevention and Treatment, School of Fundamental Medicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Yujie Zhu
- Institute of Wound Prevention and Treatment, School of Fundamental Medicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Izmailova Regina
- Institute of Wound Prevention and Treatment, School of Fundamental Medicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Yong Bi
- Department of Neurology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Fang Tong
- Institute of Wound Prevention and Treatment, School of Fundamental Medicine, Shanghai University of Medicine & Health Sciences, Shanghai, China
- Department of Neurology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| |
Collapse
|
29
|
Chen X, Soma L, Murphy C, Tretiakova M, Naresh KN, Fromm JR. Utility of CCR7 to differentiate classic Hodgkin lymphoma and other B-cell lymphomas by flow cytometry and immunohistochemistry. Am J Clin Pathol 2025; 163:266-276. [PMID: 39288406 DOI: 10.1093/ajcp/aqae119] [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: 05/06/2024] [Accepted: 08/16/2024] [Indexed: 09/19/2024] Open
Abstract
OBJECTIVES Classic Hodgkin lymphoma (CHL) is characterized by infrequent neoplastic Hodgkin and Reed-Sternberg (HRS) cells in an inflammatory background. The diagnostic utility of CC-chemokine receptor 7 (CCR7) in CHL was explored using flow cytometry and immunohistochemistry (IHC). METHODS Neoplastic specimens and non-neoplastic lymph nodes were immunophenotyped and CCR7 expression was measured semiquantitatively by flow cytometry (clone 3D12) and IHC (clone 150503). RESULTS Our results showed that CCR7 was expressed on HRS cells in the vast majority of CHL cases (45/48 by flow cytometry, 57/59 by IHC) but rarely expressed in neoplastic cells in diffuse large B-cell lymphoma, not otherwise specified (1/25 by flow cytometry, 2/40 by IHC) and nodular lymphocyte predominant Hodgkin lymphoma (0/4 by flow cytometry, 1/13 by IHC). Primary mediastinal large B-cell lymphoma (PMLBCL) revealed weak CCR7 expression by flow cytometry in most cases (8/10) but only occasionally by IHC (2/12). Both cases (2/2) of T-cell/histiocyte-rich large B-cell lymphoma (THRLBCL) also showed CCR7 expression detected by flow cytometry compared with IHC (0/7). The HRS cells demonstrated a greater percentage of positive cells and greater antigen intensity than the other B-cell lymphomas by IHC. The expression identified by flow cytometry in PMLBCL and THRLBCL but not by IHC suggests that there may be differences in the detection capabilities of the 2 techniques or the 2 CCR7 clones used. CONCLUSIONS The expression of CCR7 in HRS cells suggests its potential utility in differentiating CHL from other B-cell lymphomas. Incorporating CCR7 into flow cytometry and IHC panels may further enhance the diagnostic sensitivity of CHL.
Collapse
MESH Headings
- Humans
- Receptors, CCR7/metabolism
- Hodgkin Disease/diagnosis
- Hodgkin Disease/metabolism
- Immunohistochemistry
- Flow Cytometry
- Diagnosis, Differential
- Biomarkers, Tumor/metabolism
- Biomarkers, Tumor/analysis
- Middle Aged
- Male
- Female
- Aged
- Reed-Sternberg Cells/metabolism
- Reed-Sternberg Cells/pathology
- Adult
- Lymphoma, B-Cell/diagnosis
- Lymphoma, B-Cell/metabolism
- Immunophenotyping
- Aged, 80 and over
- Lymphoma, Large B-Cell, Diffuse/diagnosis
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Lymph Nodes/pathology
- Lymph Nodes/metabolism
- Young Adult
- Adolescent
Collapse
Affiliation(s)
- Xueyan Chen
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, US
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, US
| | - Lori Soma
- Department of Pathology, City of Hope, Duarte, CA, US
| | - Claire Murphy
- Pathology Consultants, PC, Eugene/Springfield Lab, Springfield, OR, US
| | - Maria Tretiakova
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, US
| | - Kikkeri N Naresh
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, US
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, US
| | - Jonathan R Fromm
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, US
| |
Collapse
|
30
|
Oh MS, Dumitras C, Salehi-Rad R, Tran LM, Krysan K, Lim RJ, Jing Z, Tappuni S, Lisberg A, Garon EB, Dubinett SM, Liu B. Characteristics of a CCL21 Gene-Modified Dendritic Cell Vaccine Utilized for a Clinical Trial in Non-Small Cell Lung Cancer. Mol Cancer Ther 2025; 24:286-298. [PMID: 39559833 PMCID: PMC11813162 DOI: 10.1158/1535-7163.mct-24-0435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 10/22/2024] [Accepted: 11/15/2024] [Indexed: 11/20/2024]
Abstract
The treatment of non-small cell lung cancer has made major strides with the use of immune checkpoint inhibitors; however, there remains a significant need for therapies that can overcome immunotherapy resistance. Dendritic cell (DC) vaccines have been proposed as a therapy that can potentially enhance the antitumor immune response. We have embarked on a phase I clinical trial of a vaccine consisting of monocyte-derived DCs (moDC) modified to express the chemokine C-C motif chemokine ligand 21 (CCL21-DC) given in combination with pembrolizumab. In this study, we report a comprehensive characterization of this CCL21-DC vaccine and interrogate the effects of multiple factors in the manufacturing process. We show that the cellular makeup of the CCL21-DC vaccine is heterogeneous because of the presence of passenger lymphocytes at a proportion that is highly variable among patients. Single-cell RNA sequencing of vaccines revealed further heterogeneity within the moDC compartment, with cells spanning a spectrum of DC phenotypes. Transduction with a CCL21-containing adenoviral vector augmented CCL21 secretion by moDCs, but otherwise had a minimal effect on vaccine characteristics. A single freeze-thaw cycle for stored vaccines was associated with minor alterations to the DC phenotype, as was the use of healthy donors rather than patient autologous blood. Our results highlight important considerations for the production of DC vaccines and identify underexplored factors that may affect their efficacy and immunologic impact.
Collapse
Affiliation(s)
- Michael S. Oh
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Camelia Dumitras
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Ramin Salehi-Rad
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Department of Medicine, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA
| | - Linh M. Tran
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA
| | - Kostyantyn Krysan
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA
| | - Raymond J. Lim
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Zhe Jing
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Shahed Tappuni
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Aaron Lisberg
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA
| | - Edward B. Garon
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA
| | - Steven M. Dubinett
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Department of Medicine, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA
| | - Bin Liu
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA
| |
Collapse
|
31
|
Wang Y, Li R, Tong R, Chen T, Sun M, Luo L, Li Z, Chen Y, Zhao Y, Zhang C, Wei L, Lin W, Chen H, Qian K, Chen AF, Liu J, Chen L, Li B, Wang F, Wang L, Su B, Pu J. Integrating single-cell RNA and T cell/B cell receptor sequencing with mass cytometry reveals dynamic trajectories of human peripheral immune cells from birth to old age. Nat Immunol 2025; 26:308-322. [PMID: 39881000 PMCID: PMC11785523 DOI: 10.1038/s41590-024-02059-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 12/09/2024] [Indexed: 01/31/2025]
Abstract
A comprehensive understanding of the evolution of the immune landscape in humans across the entire lifespan at single-cell transcriptional and protein levels, during development, maturation and senescence is currently lacking. We recruited a total of 220 healthy volunteers from the Shanghai Pudong Cohort (NCT05206643), spanning 13 age groups from 0 to over 90 years, and profiled their peripheral immune cells through single-cell RNA-sequencing coupled with single T cell and B cell receptor sequencing, high-throughput mass cytometry, bulk RNA-sequencing and flow cytometry validation experiments. We revealed that T cells were the most strongly affected by age and experienced the most intensive rewiring in cell-cell interactions during specific age. Different T cell subsets displayed different aging patterns in both transcriptomes and immune repertoires; examples included GNLY+CD8+ effector memory T cells, which exhibited the highest clonal expansion among all T cell subsets and displayed distinct functional signatures in children and the elderly; and CD8+ MAIT cells, which reached their peaks of relative abundance, clonal diversity and antibacterial capability in adolescents and then gradually tapered off. Interestingly, we identified and experimentally verified a previously unrecognized 'cytotoxic' B cell subset that was enriched in children. Finally, an immune age prediction model was developed based on lifecycle-wide single-cell data that can evaluate the immune status of healthy individuals and identify those with disturbed immune functions. Our work provides both valuable insights and resources for further understanding the aging of the immune system across the whole human lifespan.
Collapse
MESH Headings
- Humans
- Aged
- Single-Cell Analysis/methods
- Adult
- Aged, 80 and over
- Middle Aged
- Infant
- Child
- Child, Preschool
- Male
- Female
- Adolescent
- Aging/immunology
- Aging/genetics
- Infant, Newborn
- Receptors, Antigen, B-Cell/genetics
- Receptors, Antigen, B-Cell/immunology
- Flow Cytometry
- Young Adult
- Receptors, Antigen, T-Cell/genetics
- Sequence Analysis, RNA
- T-Lymphocyte Subsets/immunology
- B-Lymphocytes/immunology
Collapse
Affiliation(s)
- Yufei Wang
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Ronghong Li
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Renyang Tong
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Taiwei Chen
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Mingze Sun
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Lingjie Luo
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng Li
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Yifan Chen
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Yichao Zhao
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Chensheng Zhang
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Lin Wei
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Lin
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Haoyan Chen
- Department of Gastroenterology and Hepatology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kun Qian
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
- School of Biomedical Engineering, Institute of Medical Robotics and Shanghai Academy of Experimental Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Alex F Chen
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junling Liu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Chen
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Bin Li
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Wang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bing Su
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Pu
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China.
- Aging Biomarker Consortium, Beijing, China.
| |
Collapse
|
32
|
Jiang L, Duan R, Yu X, Huang Z, Peng X, Wang T, Li Z, Liu X, Wang M, Su W. An analysis of single-cell data reveals therapeutic effects of AMG487 in experimental autoimmune uveitis. Biochem Pharmacol 2025; 232:116671. [PMID: 39615601 DOI: 10.1016/j.bcp.2024.116671] [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: 07/19/2024] [Revised: 10/22/2024] [Accepted: 11/25/2024] [Indexed: 12/06/2024]
Abstract
Uveitis, an ocular autoimmune disease that poses a significant threat to vision, is caused by immune cells erroneously attacking retinal cells and currently lacks specific and effective therapeutic interventions. The CXC chemokine receptor 3 (CXCR3) facilitates the migration of immune cells to sites of inflammation. AMG487, a CXCR3 antagonist, holds potential for treating autoimmune diseases by blocking immunes cells chemotaxis. However, its effects and mechanisms in uveitis remain unclear. Using single-cell assay for transposase-accessible chromatin sequencing and RNA sequencing, we observed increased expression of CXCR3 and chemotactic pathways in peripheral blood of Vogt-Koyanagi-Harada patients and cervical lymph nodes of experimental autoimmune uveitis mice. AMG487 treatment in experimental autoimmune uveitis was shown to be therapeutically effective. Analysis of flow cytometry and single-cell RNA sequencing in AMG487-treated mice revealed reduced expression of inflammatory genes in immune cells. Specifically, AMG487 decreased the proportion of plasma cell in B cells, restored the ratio between effector T cells and regulatory T cells, and diminished T helper (Th) 17 cell pathogenicity by suppressing highly inflammatory granulocyte-macrophage colony-stimulating factor-producing Th17 cells while enhancing anti-inflammatory interleukin-10-producing Th17 cells. Our study presents an exhaustive single-cell transcriptional analysis of immune cells under AMG487 treatment, thereby elucidating potential mechanisms and providing a potential reference for the development of novel therapeutic strategies for autoimmune diseases.
Collapse
Affiliation(s)
- Loujing Jiang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Runping Duan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Xiaoyang Yu
- Guangzhou University of Chinese Medicine, Guangzhou 510060, China
| | - Zhaohao Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Xuening Peng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Tianfu Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Zhaohuai Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Xiuxing Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Mingwei Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Wenru Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China; Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
33
|
Bosteels V, Janssens S. Striking a balance: new perspectives on homeostatic dendritic cell maturation. Nat Rev Immunol 2025; 25:125-140. [PMID: 39289483 DOI: 10.1038/s41577-024-01079-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2024] [Indexed: 09/19/2024]
Abstract
Dendritic cells (DCs) are crucial gatekeepers of the balance between immunity and tolerance. They exist in two functional states, immature or mature, that refer to an information-sensing versus an information-transmitting state, respectively. Historically, the term DC maturation was used to describe the acquisition of immunostimulatory capacity by DCs following their triggering by pathogens or tissue damage signals. As such, immature DCs were proposed to mediate tolerance, whereas mature DCs were associated with the induction of protective T cell immunity. Later studies have challenged this view and unequivocally demonstrated that two distinct modes of DC maturation exist, homeostatic and immunogenic DC maturation, each with a distinct functional outcome. Therefore, the mere expression of maturation markers cannot be used to predict immunogenicity. How DCs become activated in homeostatic conditions and maintain tolerance remains an area of intense debate. Several recent studies have shed light on the signals driving the homeostatic maturation programme, especially in the conventional type 1 DC (cDC1) compartment. Here, we highlight our growing understanding of homeostatic DC maturation and the relevance of this process for immune tolerance.
Collapse
Affiliation(s)
- Victor Bosteels
- Laboratory for ER Stress and Inflammation, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Sophie Janssens
- Laboratory for ER Stress and Inflammation, VIB Center for Inflammation Research, Ghent, Belgium.
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium.
| |
Collapse
|
34
|
Wang J, Tao Q, Huang K, Wang Y, Hu L, Ren A, Wang H, Wan Y, Li J, Yi L, Ruan Y, Wanyan Z, Wu F, Zhai Z, Liu C. Chemotherapy-induced cellular senescence promotes stemness of aggressive B-cell non-Hodgkin's lymphoma via CCR7/ARHGAP18/IKBα signaling activation. J Immunother Cancer 2025; 13:e009356. [PMID: 39773566 PMCID: PMC11749403 DOI: 10.1136/jitc-2024-009356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 11/23/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Resistance to existing therapies is a major cause of treatment failure in patients with refractory and relapsed B-cell non-Hodgkin's lymphoma (r/r B-NHL). Therapy-induced senescence (TIS) is one of the most important mechanisms of drug resistance. METHODS This study used single-cell RNA sequencing to analyze doxorubicin-induced senescent B-NHL cells. C-C chemokine receptor 7 (CCR7) expression in patients with aggressive B-NHL was assessed using immunohistochemistry and flow cytometry. Lentiviral transfection was used to target CCR7 expression in Raji and SU-DHL-2 cells. Protein localization was visualized through immunofluorescence, while western blotting and co-immunoprecipitation were used to analyze protein expression and interactions. Cell proliferation was measured with the Cell Counting Kit-8 assay, and senescent cells were detected using senescence-associated β-galactosidase staining. The stemness of cells was evaluated through colony and sphere formation assays. Transwell assays assessed cell migration and invasion. Finally, inhibitors GS143 and Y27632 were used to examine the effect of IKBα and ARHGAP/RhoA inhibition on B-NHL-TIS. RESULTS Here we identified a distinct group of TIS, composed of memory B-cell population characterized by strong positive expression of CCR7, which was significantly elevated in TIS population compared with normal proliferating and autonomously senescent lymphoma cell populations. Additionally, CCR7 expression was significantly upregulated in patients with r/r B-NHL, and was an independent prognostic factor in B-NHL, with high CCR7 expression being strongly associated with poor prognosis. In vitro results indicated that CCL21 induced migration and invasion of B-NHL cells via CCR7, while blocking CCR7 reduced doxorubicin-induced migration and invasion of these cells. Furthermore, B-NHL-TIS regulated by CCR7 and exhibited enhanced phenotypic and functional stemness features, including the upregulation of stemness markers, increased colony-forming, invasive and migratory capabilities. Mechanistically, blocking CCR7 reversed the stemness characteristics of senescent B-NHL cells by inhibiting the activation of ARHGAP18/IKBα signaling. CONCLUSIONS Together, TIS promotes the stemness of B-NHL cells via CCR7/ARHGAP18/IKBα signaling activation and targeting CCR7/ARHGAP18 might overcome the chemoresistance of senescent B-NHL cells by inhibiting stemness acquisition and maintenance.
Collapse
Affiliation(s)
- Jiyu Wang
- Department of Hematology, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qianshan Tao
- Department of Hematology, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Keke Huang
- Department of Internal Medicine, The University of Hong Kong Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Yangyang Wang
- Department of Hematology, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Linhui Hu
- Department of Hematology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Anwen Ren
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Huiping Wang
- Department of Hematology, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yang Wan
- Department of Hematology, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jinlan Li
- Department of Hematology, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Liuying Yi
- Department of Hematology, The Fourth Affiliated Hospital Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Yanjie Ruan
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Zhixiang Wanyan
- Department of Emergency, The Third People's Hospital of Hefei, Hefei, Anhui, China
| | - Fan Wu
- Department of Hematology, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Zhimin Zhai
- Department of Hematology, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Chaohong Liu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
35
|
Chen W, Zhang L, Gao M, Zhang N, Wang R, Liu Y, Niu Y, Jia L. Role of tertiary lymphoid structures and B cells in clinical immunotherapy of gastric cancer. Front Immunol 2025; 15:1519034. [PMID: 39840050 PMCID: PMC11747648 DOI: 10.3389/fimmu.2024.1519034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 12/16/2024] [Indexed: 01/23/2025] Open
Abstract
Gastric cancer is a common malignant tumor of the digestive tract, and its treatment remains a significant challenge. In recent years, the role of various immune cells in the tumor microenvironment in cancer progression and treatment has gained increasing attention. Immunotherapy, primarily based on immune checkpoint inhibitors, has notably improved the prognosis of patients with gastric cancer; however, challenges regarding therapeutic efficacy persist. Histological features within the tumor microenvironment, such as tertiary lymphoid structures (TLSs), tumor-infiltrating lymphocytes, and the proportion of intratumoral stroma, are emerging as potentially effective prognostic factors. In gastric cancer, TLSs may serve as local immune hubs, enhancing the ability of immune cells to interact with and recognize tumor antigens, which is closely linked to the effectiveness of immunotherapy and improved survival rates in patients. However, the specific cell type driving TLS formation in tumors has not yet been elucidated. Mature TLSs are B-cell regions containing germinal centers. During germinal center formation, B cells undergo transformations to become mature cells with immune function, exerting anti-tumor effects. Therefore, targeting B cells within TLSs could provide new avenues for gastric cancer immunotherapy. This review, combined with current research on TLSs and B cells in gastric cancer, elaborates on the relationship between TLSs and B cells in the prognosis and immunotherapy of patients with gastric cancer, aiming to provide effective guidance for precise immunotherapy.
Collapse
Affiliation(s)
- Weiyi Chen
- Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Lingli Zhang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Man Gao
- Bayannur Clinical Medical College, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Ning Zhang
- Central Laboratory, Bayannur Hospital, Bayannur, Inner Mongolia, China
| | - Rumeng Wang
- Central Laboratory, Bayannur Hospital, Bayannur, Inner Mongolia, China
| | - Yang Liu
- Central Laboratory, Bayannur Hospital, Bayannur, Inner Mongolia, China
| | - Yan Niu
- Medical Experiment Center, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Lizhou Jia
- Central Laboratory, Bayannur Hospital, Bayannur, Inner Mongolia, China
- Medical Experiment Center, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| |
Collapse
|
36
|
Golzari-Sorkheh M, Yoganathan K, Chen ELY, Singh J, Zúñiga-Pflücker JC. T Cell Development: From T-Lineage Specification to Intrathymic Maturation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1471:81-137. [PMID: 40067585 DOI: 10.1007/978-3-031-77921-3_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2025]
Abstract
T cell development occurs in the thymus in both mice and humans. Upon entry into the thymus, bone marrow-derived blood-borne progenitors receive instructive signals, including Notch signaling, to eliminate their potential to develop into alternative immune lineages while committing to the T cell fate. Upon T-lineage commitment, developing T cells receive further instructional cues to generate different T cell sublineages, which together possess diverse immunological functions to provide host immunity. Over the years, numerous studies have contributed to a greater understanding of key thymic signals that govern T cell differentiation and subset generation. Here, we review these critical signaling factors that govern the different stages of both mouse and human T cell development, while also focusing on the transcriptional changes that mediate T cell identity and diversity.
Collapse
Affiliation(s)
- Mahdieh Golzari-Sorkheh
- Department of Immunology, University of Toronto & Sunnybrook Research Institute, Toronto, ON, Canada
| | - Kogulan Yoganathan
- Department of Immunology, University of Toronto & Sunnybrook Research Institute, Toronto, ON, Canada
| | - Edward L Y Chen
- Department of Immunology, University of Toronto & Sunnybrook Research Institute, Toronto, ON, Canada
| | - Jastaranpreet Singh
- Department of Immunology, University of Toronto & Sunnybrook Research Institute, Toronto, ON, Canada
| | | |
Collapse
|
37
|
Oh T, Woo Y, Kim G, Koo BS, Baek SH, Hwang EH, An YJ, Kim Y, Kim DY, Hong JJ. Spatiotemporal Cellular Dynamics of Germinal Center Reaction in Coronavirus Disease 2019 Lung-Draining Lymph Node Based on Imaging-Based Spatial Transcriptomics. J Transl Med 2025; 105:102180. [PMID: 39522760 DOI: 10.1016/j.labinv.2024.102180] [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: 07/03/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Although lymph node structures may be compromised in severe SARS-CoV-2 infection, the extent and parameters of recovery in convalescing patients remain unclear. Therefore, this study aimed to elucidate the nuances of lymphoid structural recovery and their implications for immunologic memory in nonhuman primates infected with SARS-CoV-2. To do so, we utilized imaging-based spatial transcriptomics to delineate immune cell composition and tissue architecture formation in the lung-draining lymph nodes during primary infection, convalescence, and reinfection from COVID-19. We noted the establishment of a germinal center with memory B cell differentiation within lymphoid follicles during convalescence accompanied by contrasting transcriptome patterns indicative of the acquisition of follicular helper T cells versus the loss of regulatory T cells. Additionally, repopulation of germinal center-like B cells was observed in the medullary niche with accumulating plasma cells along with enhanced transcriptional expression of B cell-activating factor receptor over the course of reinfection. The spatial transcriptome atlas produced herein enhances our understanding of germinal center formation with immune cell dynamics during COVID-19 convalescence and lymphoid structural recovery with transcriptome dynamics following reinfection. These findings have the potential to inform the optimization of vaccine strategies and the development of precise therapeutic interventions in the spatial context.
Collapse
Affiliation(s)
- Taehwan Oh
- National Primate Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Chungcheongbuk, Republic of Korea
| | - YoungMin Woo
- National Primate Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Chungcheongbuk, Republic of Korea; KRIBB School of Bioscience, Korea University of Science & Technology (UST), Daejeon, Republic of Korea
| | - Green Kim
- National Primate Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Chungcheongbuk, Republic of Korea
| | - Bon-Sang Koo
- National Primate Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Chungcheongbuk, Republic of Korea
| | - Seung Ho Baek
- National Primate Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Chungcheongbuk, Republic of Korea
| | - Eun-Ha Hwang
- National Primate Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Chungcheongbuk, Republic of Korea
| | - You Jung An
- National Primate Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Chungcheongbuk, Republic of Korea
| | - Yujin Kim
- National Primate Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Chungcheongbuk, Republic of Korea
| | - Dong-Yeon Kim
- National Primate Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Chungcheongbuk, Republic of Korea
| | - Jung Joo Hong
- National Primate Research Centre, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Chungcheongbuk, Republic of Korea; KRIBB School of Bioscience, Korea University of Science & Technology (UST), Daejeon, Republic of Korea.
| |
Collapse
|
38
|
Giudice GC, Sonpavde GP. Vaccine approaches to treat urothelial cancer. Hum Vaccin Immunother 2024; 20:2379086. [PMID: 39043175 PMCID: PMC11268260 DOI: 10.1080/21645515.2024.2379086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 07/08/2024] [Indexed: 07/25/2024] Open
Abstract
Bladder cancer (BC) accounts for about 4% of all malignancies. Non-muscle-invasive BC, 75% of cases, is treated with transurethral resection and adjuvant intravesical instillation, while muscle-invasive BC warrants cisplatin-based perioperative chemotherapy. Although immune-checkpoint inhibitors, antibody drug conjugates and targeted agents have provided dramatic advances, metastatic BC remains a generally incurable disease and clinical trials continue to vigorously evaluate novel molecules. Cancer vaccines aim at activating the patient's immune system against tumor cells. Several means of delivering neoantigens have been developed, including peptides, antigen-presenting cells, virus, or nucleic acids. Various improvements are constantly being explored, such as adjuvants use and combination strategies. Nucleic acids-based vaccines are increasingly gaining attention in recent years, with promising results in other malignancies. However, despite the recent advantages, numerous obstacles persist. This review is aimed at describing the different types of cancer vaccines, their evaluations in UC patients and the more recent innovations in this field.
Collapse
Affiliation(s)
- Giulia Claire Giudice
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Guru P. Sonpavde
- AdventHealth Cancer Institute, University of Central Florida, Orlando, FL, USA
| |
Collapse
|
39
|
Mundry CS, Triplett AA, Shah OS, Chaitankar V, McAndrews KL, Ly QP, Cox JL, Eberle KC, Mehla K, Swanson BJ, Lazenby A, Klute KA, Grandgenett PM, Hollingsworth MA. Single-cell RNA-sequencing of human spleens reveals an IDO-1 + tolerogenic dendritic cell subset in pancreatic cancer patients that is absent in normal individuals. Cancer Lett 2024; 607:217321. [PMID: 39547331 DOI: 10.1016/j.canlet.2024.217321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/18/2024] [Accepted: 10/31/2024] [Indexed: 11/17/2024]
Abstract
Local and systemic immunosuppression are prominent features of pancreatic cancer, rendering anti-tumor effector cells inactive and immunotherapeutic approaches ineffective. The spleen, an understudied point of antigen-presentation and T cell priming in humans, holds particular importance in pancreatic cancer due to its proximity to the developing tumor. As main effectors of antigen presentation, dendritic cells display antigens to lymphocytes, thereby bridging the innate and adaptive immune response. While tumor-infiltrating anti-inflammatory dendritic cells have been described, splenic dendritic cells have historically just been considered to stimulate the anti-tumor immune response. Here, we describe, for the first time, the presence of an immunosuppressive, tolerogenic IDO1+ dendritic cell subset in the spleens of pancreatic cancer patients that likely contributes to systemic immunosuppression that is associated with pancreatic ductal adenocarcinoma. Network analysis of scRNA seq data reveals extensive communication networks between the identified tolerogenic DC cluster and numerous immune cell populations in the spleen. Interactions with innate and adaptive immune cells suggest a broad influence on leukocyte trafficking and immune regulation within the spleen microenvironment. The identification of signaling pathways involving AHR and IDO-1, CCL19, NECTIN2, CLEC2D, and others elucidates potential mechanisms underlying the immunosuppressive functions of this cell type.
Collapse
Affiliation(s)
- Clara S Mundry
- The Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Aleata A Triplett
- The Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Osama Shiraz Shah
- Department of Internal Medicine, Division of Oncology and Hematology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Vijender Chaitankar
- Department of Internal Medicine, Division of Oncology and Hematology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kyle L McAndrews
- The Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Quan P Ly
- Department of Surgery, Division of Surgical Oncology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jesse L Cox
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kirsten C Eberle
- The Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kamiya Mehla
- Department of Oncology Science, OU Health Stephenson Cancer Center, Oklahoma City, OK, USA
| | - Benjamin J Swanson
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Audrey Lazenby
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kelsey A Klute
- Department of Internal Medicine, Division of Oncology and Hematology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Paul M Grandgenett
- The Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Michael A Hollingsworth
- The Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
40
|
Iida Y, Harada M. Local cell therapy using CCL19-expressing allogeneic mesenchymal stem cells exerts robust antitumor effects by accumulating CD103 + IL-12-producing dendritic cells and priming CD8 + T cells without involving draining lymph nodes. J Immunother Cancer 2024; 12:e009683. [PMID: 39672553 DOI: 10.1136/jitc-2024-009683] [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] [Accepted: 11/23/2024] [Indexed: 12/15/2024] Open
Abstract
BACKGROUND Immune checkpoint blockade is a promising anticancer therapy, whereas the presence of T cells in tumor sites is indispensable for its therapeutic efficacy. To promote the infiltration of T cells and dendritic cells (DCs) into the tumor, we previously proposed a local cell therapy using chemokine (C-C motif) ligand 19 (CCL19)-expressing immortalized syngeneic immortalized mesenchymal stem cells (syn-iMSC/CCL19). However, the preparation of syngeneic/autologous MSC from individual hosts limits the clinical application of this cell therapy. METHODS In this study, we further developed a new cell therapy using allogeneic iMSC/CCL19 (allo-iMSC/CCL19) using several tumor mice models. RESULTS The allo-iMSC/CCL19 therapy exerted drastic antitumor effects, in which the host's T cells were induced to respond to allogeneic MSC. In addition, the allo-iMSC/CCL19 therapy promoted the infiltration of CD103+ interleukin (IL)-12-producing DCs and priming of CD8+ T cells at tumor sites compared with that using syn-iMSC/CCL19. The antitumor effect of allo-iMSC/CCL19 therapy was not influenced by fingolimod, a sphingosine 1-phosphate receptor modulator, implying no involvement of draining lymph nodes in the priming of tumor-specific T cells. CONCLUSION These results suggest that allo-iMSC/CCL19 therapy exerts dramatic antitumor effects by promoting the infiltration of CD103+ IL-12-producing DCs and thereby priming tumor-specific CD8+ T cells at tumor sites. This local cell therapy could be a promising approach to anticancer therapy, particularly for overcoming dysfunction in the cancer-immunity cycle.
Collapse
Affiliation(s)
- Yuichi Iida
- Immunology, Shimane University Faculty of Medicine Graduate School of Medicine, Izumo, Japan
| | - Mamoru Harada
- Immunology, Shimane University Faculty of Medicine Graduate School of Medicine, Izumo, Japan
| |
Collapse
|
41
|
da Silva CP, Silva MDS, Santana HM, Paloschi MV, Ferreira E Ferreira AA, Brilhante LMV, Cruz LF, Serrath SN, Eulálio MDMC, Setúbal SDS, Vallochi AL, Nery NM, Zuliani JP. Bothrops atrox snake venom decreased MHC-II and CD86 expression in bone marrow-derived dendritic cells. Acta Trop 2024; 260:107426. [PMID: 39393479 DOI: 10.1016/j.actatropica.2024.107426] [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: 08/11/2024] [Revised: 10/05/2024] [Accepted: 10/06/2024] [Indexed: 10/13/2024]
Abstract
The effect of Bothrops atrox venom (BaV) on the maturation of bone marrow-derived dendritic cells (BMDCs) from mice was investigated, with a focus on selected cell markers, TAP1 expression, and the release of pro-inflammatory cytokines during this process. The objective was to evaluate BaV's impact on dendritic cell (DC) function, as DCs are pivotal in antigen presentation and responsible for initiating the immune response mediated by naïve T cells, as well as regulating the immune system. Bone marrow cells were obtained from Swiss mice, and hematopoietic precursors were differentiated into BMDCs using GM-CSF and IL-4. On the 7th day, BaV and LPS were introduced into the culture, and the cells were analyzed 24 h later. BaV's ability to stimulate BMDC maturation was assessed through the analysis of surface marker expression. The findings demonstrated that BMDCs are highly influenced by culture environment factors, such as GM-CSF and IL-4, and are sensitive to additional stimuli like LPS and BaV. Mature DCs exhibited elevated levels of critical markers for T cell activation, such as MHC-II, CD80, and CD86, displaying specific phenotypic characteristics. However, the observed reduction in MHC-II and CD86 expression following BaV exposure suggests a substantial impact on the immunological activation capacity of these cells, potentially interfering with the adaptive immune response. Furthermore, the selective release of cytokines, such as IL-6, but not TNF-α or IL-1β, indicates differentiated modulation of inflammatory responses by DCs under various stimulation conditions.
Collapse
Affiliation(s)
- Carolina P da Silva
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Milena D S Silva
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Hallison M Santana
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Mauro V Paloschi
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Alex A Ferreira E Ferreira
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Lívia M V Brilhante
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Larissa F Cruz
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Suzanne N Serrath
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Micaela de M C Eulálio
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Sulamita da S Setúbal
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Adriana L Vallochi
- Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, RJ, Brazil
| | - Neriane M Nery
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil.
| | - Juliana P Zuliani
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil; Departamento de Medicina, Universidade Federal de Rondônia, UNIR, Porto Velho, RO, Brazil.
| |
Collapse
|
42
|
Scholaert M, Peries M, Braun E, Martin J, Serhan N, Loste A, Bruner A, Basso L, Chaput B, Merle E, Descargues P, Pagès E, Gaudenzio N. Multimodal profiling of biostabilized human skin modules reveals a coordinated ecosystem response to injected mRNA-1273 COVID-19 vaccine. Allergy 2024; 79:3341-3359. [PMID: 39157907 PMCID: PMC11657073 DOI: 10.1111/all.16273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND The field of drug development is witnessing a remarkable surge in the development of innovative strategies. There is a need to develop technological platforms capable of generating human data prior to progressing to clinical trials. METHODS Here we introduce a new flexible solution designed for the comprehensive monitoring of the natural human skin ecosystem's response to immunogenic drugs over time. Based on unique bioengineering to preserve surgical resections in a long survival state, it allows for the first time a comprehensive analysis of resident immune cells response at both organ and single-cell levels. RESULTS Upon injection of the mRNA-1273 COVID-19 vaccine, we characterized precise sequential molecular events triggered upon detection of the exogenous substance. The vaccine consistently targets DC/macrophages and mast cells, regardless of the administration route, while promoting specific cell-cell communications in surrounding immune cell subsets. CONCLUSION Given its direct translational relevance, this approach provides a multiscale vision of genuine human tissue immunity that could pave the way toward the development of new vaccination and drug development strategies.
Collapse
Affiliation(s)
- Manon Scholaert
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERM UMR1291—CNRS UMR5051University Toulouse IIIToulouseFrance
- Genoskin SASToulouseFrance
| | | | | | - Jeremy Martin
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERM UMR1291—CNRS UMR5051University Toulouse IIIToulouseFrance
| | - Nadine Serhan
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERM UMR1291—CNRS UMR5051University Toulouse IIIToulouseFrance
| | - Alexia Loste
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERM UMR1291—CNRS UMR5051University Toulouse IIIToulouseFrance
| | - Audrey Bruner
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERM UMR1291—CNRS UMR5051University Toulouse IIIToulouseFrance
| | - Lilian Basso
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERM UMR1291—CNRS UMR5051University Toulouse IIIToulouseFrance
| | - Benoît Chaput
- Department of Plastic, Reconstructive and Aesthetic Surgery, Rangueil HospitalCHU ToulouseToulouseFrance
| | | | | | | | - Nicolas Gaudenzio
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERM UMR1291—CNRS UMR5051University Toulouse IIIToulouseFrance
- Genoskin SASToulouseFrance
| |
Collapse
|
43
|
Zhang J, Guo T, Chen Y, Wang X, Wu L, Xie H. Investigating the causal relationship between inflammation and multiple types of hearing loss: a multi-omics approach combining Mendelian randomization and molecular docking. Front Neurol 2024; 15:1422241. [PMID: 39677857 PMCID: PMC11638537 DOI: 10.3389/fneur.2024.1422241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 11/08/2024] [Indexed: 12/17/2024] Open
Abstract
Background Hearing loss affects over 10% of the global population. Inflammation is a key factor in hearing loss caused by noise, infection, and aging, damaging various hearing-related tissues (e.g., spiral ligament, stria vascularis). Mendelian randomization (MR) can help identify potential causal relationships and therapeutic targets. Methods We conducted MR analyses on 91 inflammatory proteins (n = 14,824) and genome-wide association study results for various hearing loss types in European ancestry populations, including sensorineural hearing loss (SNHL; ncases = 15,952, ncontrols = 196,592), sudden idiopathic hearing loss (SIHL; ncases = 1,491, ncontrols = 196,592), and other hearing loss (OHL; ncases = 4,157, ncontrols = 196,592). Additionally, hearing loss with difficulty in hearing (ncases = 14,654, ncontrols = 474,839) served as a validation set. To predict inflammatory protein-enriched pathways and tissues, we performed enrichment analysis, functional annotation, and tissue analyses using "OmicsNet2.0" and "FUMA" platforms. We also combined "CoreMine" and molecular docking to explore potential drugs targeting inflammatory proteins and investigate binding efficacy. Results CCL19 was identified as a common risk factor for SNHL and OHL, which was validated in the hearing loss with difficulty in hearing dataset. Tissue analysis revealed that SIHL-related inflammatory proteins were enriched in the amygdala. Multi-omics research indicated associations between inflammatory proteins and neurodegenerative diseases. Molecular docking studies suggested that Chuanxiong Rhizoma and Uncariae Ramulus Cumuncis are potential drugs for targeting CCL19. Conclusion This study identified CCL19 as a common risk factor for various types of hearing loss through MR analysis, highlighting the crucial role of inflammatory proteins in hearing loss. The enrichment of related inflammatory proteins in the amygdala and their association with neurodegenerative diseases provide new insights into the mechanisms of hearing loss.
Collapse
Affiliation(s)
- Jingqi Zhang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tao Guo
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yaxin Chen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiangjin Wang
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, China
| | - Lijiao Wu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hui Xie
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
44
|
Kadam R, Gupta M, Lazarov O, Prabhakar BS. Brain-immune interactions: implication for cognitive impairments in Alzheimer's disease and autoimmune disorders. J Leukoc Biol 2024; 116:1269-1290. [PMID: 38869088 DOI: 10.1093/jleuko/qiae134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 05/09/2024] [Accepted: 06/11/2024] [Indexed: 06/14/2024] Open
Abstract
Progressive memory loss and cognitive dysfunction, encompassing deficits in learning, memory, problem solving, spatial reasoning, and verbal expression, are characteristics of Alzheimer's disease and related dementia. A wealth of studies has described multiple roles of the immune system in the development or exacerbation of dementia. Individuals with autoimmune disorders can also develop cognitive dysfunction, a phenomenon termed "autoimmune dementia." Together, these findings underscore the pivotal role of the neuroimmune axis in both Alzheimer's disease and related dementia and autoimmune dementia. The dynamic interplay between adaptive and innate immunity, both in and outside the brain, significantly affects the etiology and progression of these conditions. Multidisciplinary research shows that cognitive dysfunction arises from a bidirectional relationship between the nervous and immune systems, though the specific mechanisms that drive cognitive impairments are not fully understood. Intriguingly, this reciprocal regulation occurs at multiple levels, where neuronal signals can modulate immune responses, and immune system-related processes can influence neuronal viability and function. In this review, we consider the implications of autoimmune responses in various autoimmune disorders and Alzheimer's disease and explore their effects on brain function. We also discuss the diverse cellular and molecular crosstalk between the brain and the immune system, as they may shed light on potential triggers of peripheral inflammation, their effect on the integrity of the blood-brain barrier, and brain function. Additionally, we assess challenges and possibilities associated with developing immune-based therapies for the treatment of cognitive decline.
Collapse
Affiliation(s)
- Rashmi Kadam
- Department of Microbiology and Immunology, University of Illinois College of Medicine, 835 S Wolcott street, MC 790, Chicago, Chicago, IL 60612, United States
| | - Muskan Gupta
- Department of Anatomy and Cell Biology, University of Illinois College of Medicine, 808 S Wood street, MC 512, Chicago, Chicago, IL 60612, United States
| | - Orly Lazarov
- Department of Anatomy and Cell Biology, University of Illinois College of Medicine, 808 S Wood street, MC 512, Chicago, Chicago, IL 60612, United States
| | - Bellur S Prabhakar
- Department of Microbiology and Immunology, University of Illinois College of Medicine, 835 S Wolcott street, MC 790, Chicago, Chicago, IL 60612, United States
| |
Collapse
|
45
|
Zhang H, Luo M, Li Y, Liu L, Bian J, Gong L, He C, Han L, Wang M. Ellagic acid ameliorates alcohol-induced cognitive and social dysfunction through the gut microbiota-mediated CCL21-CCR7 axis. Food Funct 2024; 15:11186-11205. [PMID: 39449276 DOI: 10.1039/d4fo03985h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
Chronic alcohol consumption disrupts the balance of the gut microbiome, resulting in alcohol-induced cognitive and social dysfunction (AICSD), and serves as a primary etiological factor for early-onset dementia. Ellagic acid (EA) is a polyphenolic compound belonging to the ellagitannin family, showing potential as a dietary intervention for alleviating cognitive impairments. Nonetheless, the protective effects and underlying mechanisms of EA on AICSD remain unclear. In our study, we employed a multi-omics approach to elucidate the microbiome-mediated mechanism underlying the beneficial effects of EA on AICSD. Firstly, our findings demonstrate that EA significantly ameliorated cognitive and social behavioral deficits as well as neuroinflammation induced by alcohol. Moreover, RNA-seq analysis of hippocampi indicates that EA regulated the KEGG pathway of cytokine-cytokine receptor interaction signaling by downregulating the CCL21-CCR7 axis. Furthermore, we observed that EA effectively restored the dysbiosis of gut microbiota and their derived metabolites induced by chronic alcohol consumption. Strong connections were observed between EA-regulated genes, microbiota and metabolites. Finally, the causal relationship between the microbiome and behavioral changes was further confirmed through antibiotic treatment and fecal microbiota transplantation experiments. Overall, our study provides innovative evidence supporting the role of EA in improving AICSD via regulation of the cytokine-cytokine receptor interaction signaling pathway through the microbiota-mediated CCl21-CCR7 axis. These findings offer valuable insights into both EA-based interventions as well as microbial interventions against AICSD.
Collapse
Affiliation(s)
- Hongbo Zhang
- Department of Nutrition and Health, College of Food Science and Engineering, Northwest A&F University, Yang ling 712100, Shaanxi, China.
| | - Min Luo
- Department of Nutrition and Health, College of Food Science and Engineering, Northwest A&F University, Yang ling 712100, Shaanxi, China.
| | - Yinuo Li
- Department of Nutrition and Health, College of Food Science and Engineering, Northwest A&F University, Yang ling 712100, Shaanxi, China.
| | - Lu Liu
- Department of Nutrition and Health, College of Food Science and Engineering, Northwest A&F University, Yang ling 712100, Shaanxi, China.
| | - Ji Bian
- Kolling Institute, Sydney Medical School, Royal North Shore Hospital, University of Sydney, St Leonards, NSW 2065, Australia
| | - Lan Gong
- UNSW Microbiome Research Centre, St George and Sutherland Clinical Campus, University of New South Wales, Sydney, NSW 2052, Australia
| | - Caian He
- Department of Nutrition and Health, College of Food Science and Engineering, Northwest A&F University, Yang ling 712100, Shaanxi, China.
| | - Lin Han
- Department of Nutrition and Health, College of Food Science and Engineering, Northwest A&F University, Yang ling 712100, Shaanxi, China.
| | - Min Wang
- Department of Nutrition and Health, College of Food Science and Engineering, Northwest A&F University, Yang ling 712100, Shaanxi, China.
| |
Collapse
|
46
|
Niemietz P, Peiseler M, Kohlhepp M, Horn P, Matchett K, Wang Y, Haas L, Zhang T, Bruneau A, Guillot A, Berger H, Liepelt A, Warzecha K, Demske C, Möckel D, Lammers T, Henderson N, Heymann F, Tacke F. C-C chemokine receptor type 7 (CCR7) regulates hepatic CD8 + T cell homeostasis and response to acute liver injury. Hepatology 2024; 80:1104-1119. [PMID: 38231043 DOI: 10.1097/hep.0000000000000757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 12/20/2023] [Indexed: 01/18/2024]
Abstract
BACKGROUND AND AIMS Acute liver failure (ALF) is a rare but life-threatening condition, and DILI, particularly acetaminophen toxicity, is the leading cause of ALF. Innate immune mechanisms further perpetuate liver injury, while the role of the adaptive immune system in DILI-related ALF is unclear. APPROACH AND RESULTS We analyzed liver tissue from 2 independent patient cohorts with ALF and identified hepatic T cell infiltration as a prominent feature in human ALF. CD8 + T cells were characterized by zonation toward necrotic regions and an activated gene expression signature. In murine acetaminophen-induced liver injury, intravital microscopy revealed zonation of CD8 + but not CD4 + T cells at necrotic areas. Gene expression analysis exposed upregulated C-C chemokine receptor 7 (CCR7) and its ligand CCL21 in the liver as well as a broadly activated phenotype of hepatic CD8 + T cells. In 2 mouse models of ALF, Ccr7-/- mice had significantly aggravated early-phase liver damage. Functionally, CCR7 was not involved in the recruitment of CD8 + T cells, but regulated their activation profile potentially through egress to lymphatics. Ccr7-/- CD8 + T cells were characterized by elevated expression of activation, effector, and exhaustion profiles. Adoptive transfer revealed preferential homing of CCR7-deficient CD8 + T cells to the liver, and depletion of CD8 + T cells attenuated liver damage in mice. CONCLUSIONS Our study demonstrates the involvement of the adaptive immune system in ALF in humans and mice. We identify the CCR7-CCL21 axis as an important regulatory pathway, providing downstream protection against T cell-mediated liver injury.
Collapse
Affiliation(s)
- Patricia Niemietz
- Department of Medicine III, RWTH-University Hospital Aachen, Aachen, Germany
| | - Moritz Peiseler
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Marlene Kohlhepp
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| | - Paul Horn
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Kylie Matchett
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Yuting Wang
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| | - Leon Haas
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| | - Tianjiao Zhang
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| | - Alix Bruneau
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| | - Adrien Guillot
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| | - Hilmar Berger
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| | - Anke Liepelt
- Department of Medicine III, RWTH-University Hospital Aachen, Aachen, Germany
| | - Klaudia Warzecha
- Department of Medicine III, RWTH-University Hospital Aachen, Aachen, Germany
| | - Catharina Demske
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| | - Diana Möckel
- Department of Nanomedicine and Theranostics, Institute for Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Neil Henderson
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Felix Heymann
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| | - Frank Tacke
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| |
Collapse
|
47
|
Clua‐Ferré L, Suau R, Vañó‐Segarra I, Ginés I, Serena C, Manyé J. Therapeutic potential of mesenchymal stem cell-derived extracellular vesicles: A focus on inflammatory bowel disease. Clin Transl Med 2024; 14:e70075. [PMID: 39488745 PMCID: PMC11531661 DOI: 10.1002/ctm2.70075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/13/2024] [Accepted: 10/16/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) have emerged as key regulators of intercellular communication, orchestrating essential biological processes by delivering bioactive cargoes to target cells. Available evidence suggests that MSC-EVs can mimic the functions of their parental cells, exhibiting immunomodulatory, pro-regenerative, anti-apoptotic, and antifibrotic properties. Consequently, MSC-EVs represent a cell-free therapeutic option for patients with inflammatory bowel disease (IBD), overcoming the limitations associated with cell replacement therapy, including their non-immunogenic nature, lower risk of tumourigenicity, cargo specificity and ease of manipulation and storage. MAIN TOPICS COVERED This review aims to provide a comprehensive examination of the therapeutic efficacy of MSC-EVs in IBD, with a focus on their mechanisms of action and potential impact on treatment outcomes. We examine the advantages of MSC-EVs over traditional therapies, discuss methods for their isolation and characterisation, and present mechanistic insights into their therapeutic effects through transcriptomic, proteomic and lipidomic analyses of MSC-EV cargoes. We also discuss available preclinical studies demonstrating that MSC-EVs reduce inflammation, promote tissue repair and restore intestinal homeostasis in IBD models, and compare these findings with those of clinical trials. CONCLUSIONS Finally, we highlight the potential of MSC-EVs as a novel therapy for IBD and identify challenges and opportunities associated with their translation into clinical practice. HIGHLIGHTS The source of mesenchymal stem cells (MSCs) strongly influences the composition and function of MSC-derived extracellular vesicles (EVs), affecting their therapeutic potential. Adipose-derived MSC-EVs, known for their immunoregulatory properties and ease of isolation, show promise as a treatment for inflammatory bowel disease (IBD). MicroRNAs are consistently present in MSC-EVs across cell types and are involved in pathways that are dysregulated in IBD, making them potential therapeutic agents. For example, miR-let-7a is associated with inhibition of apoptosis, miR-100 supports cell survival, miR-125b helps suppress pro-inflammatory cytokines and miR-20 promotes anti-inflammatory M2 macrophage polarisation. Preclinical studies in IBD models have shown that MSC-EVs reduce intestinal inflammation by suppressing pro-inflammatory mediators (e.g., TNF-α, IL-1β, IL-6) and increasing anti-inflammatory factors (e.g., IL-4, IL-10). They also promote mucosal healing and strengthen the integrity of the gut barrier, suggesting their potential to address IBD pathology.
Collapse
Affiliation(s)
- Laura Clua‐Ferré
- Germans Trias i Pujol Research Institute IGTPInflammatory Bowel DiseasesBadalonaSpain
| | - Roger Suau
- Germans Trias i Pujol Research Institute IGTPInflammatory Bowel DiseasesBadalonaSpain
| | - Irene Vañó‐Segarra
- Hospital Universitari Joan XXIIIInstitut d'investigació sanitària Pere VirgiliTarragonaSpain
| | - Iris Ginés
- Hospital Universitari Joan XXIIIInstitut d'investigació sanitària Pere VirgiliTarragonaSpain
| | - Carolina Serena
- Hospital Universitari Joan XXIIIInstitut d'investigació sanitària Pere VirgiliTarragonaSpain
| | - Josep Manyé
- Germans Trias i Pujol Research Institute IGTPInflammatory Bowel DiseasesBadalonaSpain
- Centro de Investigación Biomédica en RedMadridSpain
| |
Collapse
|
48
|
Wang C, Macintyre AN, Oguin TH, McCarthy KR, Moody MA, Yuan F. Spatiotemporal control of immune responses with nucleic acid cocktail vaccine. ADVANCED THERAPEUTICS 2024; 7:2400263. [PMID: 40248361 PMCID: PMC12002596 DOI: 10.1002/adtp.202400263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Indexed: 04/19/2025]
Abstract
Nucleic acid vaccines play important roles in prevention and treatment of diseases. However, limited immunogenicity remains a major obstacle for DNA vaccine applications in the clinic. To address the issue, the present study investigates a cocktail approach to DNA vaccination. In this proof-of-the-concept study, the cocktail consists of two DNAs encoding viral hemagglutinin (HA) and granulocyte-macrophage colony stimulatory factor (GM-CSF), respectively. Data from the study demonstrate that recruitment and activation of antigen-presenting cells (APCs) can be substantially improved by spatiotemporal regulation of GM-CSF and HA expressions at the site of vaccination. The types of recruited APCs and their phenotypes are also controllable by adjusting the cocktail compositions. Compared to mono-ingredient vaccine, the optimized cocktail vaccine is able to enhance the anti-viral humoral and T cell immune responses. No significant systemic inflammation has been detected after either prime or boost immunization using the cocktail vaccine. Data in the study suggest that the DNA cocktail is a safe, effective, and controllable platform for improving vaccine efficacy.
Collapse
Affiliation(s)
- Chunxi Wang
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Andrew N. Macintyre
- Duke Human Vaccine Institute (DHVI), Duke University, Durham, NC 27708, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27708, USA
| | - Thomas H. Oguin
- Duke Human Vaccine Institute (DHVI), Duke University, Durham, NC 27708, USA
| | - Kevin R. McCarthy
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine; Pittsburgh, PA 15261, USA
| | - M. Anthony Moody
- Duke Human Vaccine Institute (DHVI), Duke University, Durham, NC 27708, USA
- Department of Pediatrics, Duke University School of Medicine, Durham, NC 27708, USA
| | - Fan Yuan
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| |
Collapse
|
49
|
Reddiar SB, Xie Y, Abdallah M, Han S, Hu L, Feeney OM, Gracia G, Anshabo A, Lu Z, Farooq MA, Styles IK, Phillips ARJ, Windsor JA, Porter CJH, Cao E, Trevaskis NL. Intestinal Lymphatic Biology, Drug Delivery, and Therapeutics: Current Status and Future Directions. Pharmacol Rev 2024; 76:1326-1398. [PMID: 39179383 DOI: 10.1124/pharmrev.123.001159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 07/29/2024] [Accepted: 08/14/2024] [Indexed: 08/26/2024] Open
Abstract
Historically, the intestinal lymphatics were considered passive conduits for fluids, immune cells, dietary lipids, lipid soluble vitamins, and lipophilic drugs. Studies of intestinal lymphatic drug delivery in the late 20th century focused primarily on the drugs' physicochemical properties, especially high lipophilicity, that resulted in intestinal lymphatic transport. More recent discoveries have changed our traditional view by demonstrating that the lymphatics are active, plastic, and tissue-specific players in a range of biological and pathological processes, including within the intestine. These findings have, in turn, inspired exploration of lymph-specific therapies for a range of diseases, as well as the development of more sophisticated strategies to actively deliver drugs or vaccines to the intestinal lymph, including a range of nanotechnologies, lipid prodrugs, and lipid-conjugated materials that "hitchhike" onto lymphatic transport pathways. With the increasing development of novel therapeutics such as biologics, there has been interest in whether these therapeutics are absorbed and transported through intestinal lymph after oral administration. Here we review the current state of understanding of the anatomy and physiology of the gastrointestinal lymphatic system in health and disease, with a focus on aspects relevant to drug delivery. We summarize the current state-of-the-art approaches to deliver drugs and quantify their uptake into the intestinal lymphatic system. Finally, and excitingly, we discuss recent examples of significant pharmacokinetic and therapeutic benefits achieved via intestinal lymphatic drug delivery. We also propose approaches to advance the development and clinical application of intestinal lymphatic delivery strategies in the future. SIGNIFICANCE STATEMENT: This comprehensive review details the understanding of the anatomy and physiology of the intestinal lymphatic system in health and disease, with a focus on aspects relevant to drug delivery. It highlights current state-of-the-art approaches to deliver drugs to the intestinal lymphatics and the shift toward the use of these strategies to achieve pharmacokinetic and therapeutic benefits for patients.
Collapse
Affiliation(s)
- Sanjeevini Babu Reddiar
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Yining Xie
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Mohammad Abdallah
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Sifei Han
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Luojuan Hu
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Orlagh M Feeney
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Gracia Gracia
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Abel Anshabo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Zijun Lu
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Muhammad Asim Farooq
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Ian K Styles
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Anthony R J Phillips
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - John A Windsor
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Christopher J H Porter
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Enyuan Cao
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Natalie L Trevaskis
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| |
Collapse
|
50
|
Zhao L, Shireman J, Probelsky S, Rigg B, Wang X, Huff WX, Kwon JH, Dey M. CCL21 Induces Plasmacytoid Dendritic Cell Migration and Activation in a Mouse Model of Glioblastoma. Cancers (Basel) 2024; 16:3459. [PMID: 39456552 PMCID: PMC11506458 DOI: 10.3390/cancers16203459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Dendritic cells (DCs) are professional antigen-presenting cells that are traditionally divided into two distinct subsets: myeloid DCs (mDCs) and plasmacytoid DCs (pDCs). pDCs are known for their ability to secrete large amounts of cytokine type I interferons (IFN- α). In our previous work, we have demonstrated that pDC infiltration promotes glioblastoma (GBM) tumor immunosuppression through decreased IFN-α secretion via TLR-9 signaling and increased suppressive function of regulatory T cells (Tregs) via increased IL-10 secretion, resulting in poor overall outcomes in mouse models of GBM. Further dissecting the overall mechanism of pDC-mediated GBM immunosuppression, in this study, we identified CCL21 as highly upregulated by multiple GBM cell lines, which recruit pDCs to tumor sites via CCL21-CCR7 signaling. Furthermore, pDCs are activated by CCL21 in the GBM microenvironment through intracellular signaling of β-arrestin and CIITA. Finally, we found that CCL21-treated pDCs directly suppress CD8+ T cell proliferation without affecting regulatory T cells (Tregs) differentiation, which is considered the canonical pathway of immunotolerant regulation. Taken together, our results show that pDCs play a multifaced role in GBM immunosuppression, and CCL21 could be a novel therapeutic target in GBM to overcome pDC-mediated immunosuppression.
Collapse
Affiliation(s)
- Lei Zhao
- Department of Neurosurgery, University of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center, Madison, WI 53706, USA; (L.Z.); (J.S.); (S.P.); (B.R.); (X.W.)
| | - Jack Shireman
- Department of Neurosurgery, University of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center, Madison, WI 53706, USA; (L.Z.); (J.S.); (S.P.); (B.R.); (X.W.)
| | - Samantha Probelsky
- Department of Neurosurgery, University of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center, Madison, WI 53706, USA; (L.Z.); (J.S.); (S.P.); (B.R.); (X.W.)
| | - Bailey Rigg
- Department of Neurosurgery, University of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center, Madison, WI 53706, USA; (L.Z.); (J.S.); (S.P.); (B.R.); (X.W.)
| | - Xiaohu Wang
- Department of Neurosurgery, University of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center, Madison, WI 53706, USA; (L.Z.); (J.S.); (S.P.); (B.R.); (X.W.)
| | - Wei X. Huff
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (W.X.H.); (J.H.K.)
| | - Jae H. Kwon
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (W.X.H.); (J.H.K.)
| | - Mahua Dey
- Department of Neurosurgery, University of Wisconsin School of Medicine & Public Health, UW Carbone Cancer Center, Madison, WI 53706, USA; (L.Z.); (J.S.); (S.P.); (B.R.); (X.W.)
| |
Collapse
|