1
|
Roberti MP, Charoentong P, Lyu Y, Meyer M, Eichmüller SB, Schmidt P, Momburg F, Cetin M, Hartmann F, Valous NA, Stenzinger A, Michel L, Lichter P, Schneeweiss A, Thewes V, Fremd C, Zörnig I, Jäger D. Isolation of a tumor neoantigen specific CD8+ TCR from a skin biopsy of a vaccination site. Oncoimmunology 2025; 14:2457793. [PMID: 39902862 PMCID: PMC11796541 DOI: 10.1080/2162402x.2025.2457793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 01/19/2025] [Accepted: 01/20/2025] [Indexed: 02/06/2025] Open
Abstract
T cells that recognize tumor-specific mutations are crucial for cancer immunosurveillance and in adoptive transfer of TILs or transgenic-TCR T cell products. However, their challenging identification and isolation limits their use in clinical practice. Therefore, novel approaches to isolate tumor-specific T cells are needed. Here, we report the isolation of neoantigen-specific CD8+ T cells from a vaccination site of a metastatic breast cancer patient who received a personalized vaccine. Based on the somatic mutations, potential MHC binding epitopes were predicted, of which 17 were selected to generate a peptide vaccine. Cutaneous biopsies were processed after the fifth vaccination cycle to obtain infiltrating lymphocytes from the vaccination site (VILs). IFNγ ELISpot revealed reactivity to four peptides used in the vaccine. Reactive T cells from VILs were non-overlapping with those detected in the blood and the tumor-microenvironment. ScTCR Seq analysis revealed the presence of a clonotype in VILs that further expanded after a round of in vitro stimulation and validated to be specific against a private mutation, namely NCOR1L1475R, presented in the context of HLA-B * 07:02, with no reactivity to the wild-type peptide. Our study shows, for the first time, that tumor mutation - specific T cells are generated at high frequencies in the vaccination site and can be isolated with standard methods for TCR screening. The easy and safe accessibility of skin biopsies overcomes the major hurdles of current TCR screening approaches and present exciting opportunities for the development of innovative immunotherapeutic strategies.
Collapse
Affiliation(s)
- Maria Paula Roberti
- Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Medical Faculty Heidelberg, Department of Medical Oncology and Internal Medicine VI, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, A partnership between DKFZ and Heidelberg University Medical Center, Heidelberg, Germany
| | - Pornpimol Charoentong
- Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Medical Faculty Heidelberg, Department of Medical Oncology and Internal Medicine VI, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, A partnership between DKFZ and Heidelberg University Medical Center, Heidelberg, Germany
- Center for Quantitative Analysis of Molecular and Cellular Biosystems (Bioquant), Heidelberg University, Heidelberg, Germany
| | - Yanhong Lyu
- Medical Faculty Heidelberg, Department of Medical Oncology and Internal Medicine VI, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
- Center for Quantitative Analysis of Molecular and Cellular Biosystems (Bioquant), Heidelberg University, Heidelberg, Germany
| | - Marten Meyer
- Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Medical Faculty Heidelberg, Department of Medical Oncology and Internal Medicine VI, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, A partnership between DKFZ and Heidelberg University Medical Center, Heidelberg, Germany
- Antigen Presentation and T/NK Cell Activation Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan B. Eichmüller
- GMP and T Cell Therapy, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Patrick Schmidt
- Medical Faculty Heidelberg, Department of Medical Oncology and Internal Medicine VI, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, A partnership between DKFZ and Heidelberg University Medical Center, Heidelberg, Germany
- GMP and T Cell Therapy, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Frank Momburg
- Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Medical Faculty Heidelberg, Department of Medical Oncology and Internal Medicine VI, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
- Antigen Presentation and T/NK Cell Activation Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Miray Cetin
- Systems Immunology and Single Cell Biology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Felix Hartmann
- Systems Immunology and Single Cell Biology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Core Center Heidelberg, Heidelberg, Germany
| | - Nektarios A. Valous
- Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Medical Faculty Heidelberg, Department of Medical Oncology and Internal Medicine VI, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
- Center for Quantitative Analysis of Molecular and Cellular Biosystems (Bioquant), Heidelberg University, Heidelberg, Germany
| | | | - Laura Michel
- Medical Faculty Heidelberg, Department of Medical Oncology and Internal Medicine VI, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
- Division of Gynecological Oncology, National Center for Tumor Diseases (NCT), A partnership between DKFZ and Heidelberg University Medical Center, Heidelberg University Hospital, Heidelberg, Germany
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter Lichter
- National Center for Tumor Diseases (NCT) Heidelberg, A partnership between DKFZ and Heidelberg University Medical Center, Heidelberg, Germany
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andreas Schneeweiss
- Division of Gynecological Oncology, National Center for Tumor Diseases (NCT), A partnership between DKFZ and Heidelberg University Medical Center, Heidelberg University Hospital, Heidelberg, Germany
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Verena Thewes
- National Center for Tumor Diseases (NCT) Heidelberg, A partnership between DKFZ and Heidelberg University Medical Center, Heidelberg, Germany
- Division of Gynecological Oncology, National Center for Tumor Diseases (NCT), A partnership between DKFZ and Heidelberg University Medical Center, Heidelberg University Hospital, Heidelberg, Germany
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Carlo Fremd
- Medical Faculty Heidelberg, Department of Medical Oncology and Internal Medicine VI, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
- Division of Gynecological Oncology, National Center for Tumor Diseases (NCT), A partnership between DKFZ and Heidelberg University Medical Center, Heidelberg University Hospital, Heidelberg, Germany
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Inka Zörnig
- Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Medical Faculty Heidelberg, Department of Medical Oncology and Internal Medicine VI, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, A partnership between DKFZ and Heidelberg University Medical Center, Heidelberg, Germany
| | - Dirk Jäger
- Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Medical Faculty Heidelberg, Department of Medical Oncology and Internal Medicine VI, Heidelberg University Hospital, Heidelberg University, Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, A partnership between DKFZ and Heidelberg University Medical Center, Heidelberg, Germany
| |
Collapse
|
2
|
Sparano C, Solís-Sayago D, Zangger NS, Rindlisbacher L, Van Hove H, Vermeer M, Westermann F, Mussak C, Rallo E, Dergun S, Litscher G, Xu Y, Bijnen M, Friedrich C, Greter M, Juranić Lisnić V, Becher B, Gasteiger G, Oxenius A, Tugues S. Autocrine TGF-β1 drives tissue-specific differentiation and function of resident NK cells. J Exp Med 2025; 222:e20240930. [PMID: 39692745 DOI: 10.1084/jem.20240930] [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: 05/29/2024] [Revised: 10/21/2024] [Accepted: 11/20/2024] [Indexed: 12/19/2024] Open
Abstract
Group 1 innate lymphoid cells (ILCs) encompass NK cells and ILC1s, which have non-redundant roles in host protection against pathogens and cancer. Despite their circulating nature, NK cells can establish residency in selected tissues during ontogeny, forming a distinct functional subset. The mechanisms that initiate, maintain, and regulate the conversion of NK cells into tissue-resident NK (trNK) cells are currently not well understood. Here, we identify autocrine transforming growth factor-β (TGF-β) as a cell-autonomous driver for NK cell tissue residency across multiple glandular tissues during development. Cell-intrinsic production of TGF-β was continuously required for the maintenance of trNK cells and synergized with Hobit to enhance cytotoxic function. Whereas autocrine TGF-β was redundant in tumors, our study revealed that NK cell-derived TGF-β allowed the expansion of cytotoxic trNK cells during local infection with murine cytomegalovirus (MCMV) and contributed to viral control in the salivary gland. Collectively, our findings reveal tissue-specific regulation of trNK cell differentiation and function by autocrine TGF-β1, which is relevant for antiviral immunity.
Collapse
Affiliation(s)
- Colin Sparano
- Institute of Experimental Immunology, University of Zürich , Zürich, Switzerland
| | - Darío Solís-Sayago
- Institute of Experimental Immunology, University of Zürich , Zürich, Switzerland
| | | | - Lukas Rindlisbacher
- Institute of Experimental Immunology, University of Zürich , Zürich, Switzerland
| | - Hannah Van Hove
- Institute of Experimental Immunology, University of Zürich , Zürich, Switzerland
| | - Marijne Vermeer
- Institute of Experimental Immunology, University of Zürich , Zürich, Switzerland
| | - Frederike Westermann
- Institute of Experimental Immunology, University of Zürich , Zürich, Switzerland
| | - Caroline Mussak
- Institute of Experimental Immunology, University of Zürich , Zürich, Switzerland
| | - Elisa Rallo
- Institute of Experimental Immunology, University of Zürich , Zürich, Switzerland
| | - Stanislav Dergun
- Institute of Experimental Immunology, University of Zürich , Zürich, Switzerland
| | - Gioana Litscher
- Institute of Experimental Immunology, University of Zürich , Zürich, Switzerland
| | - Yishu Xu
- Institute of Experimental Immunology, University of Zürich , Zürich, Switzerland
| | - Mitchell Bijnen
- Institute of Experimental Immunology, University of Zürich , Zürich, Switzerland
| | - Christin Friedrich
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg , Würzburg, Germany
| | - Melanie Greter
- Institute of Experimental Immunology, University of Zürich , Zürich, Switzerland
| | - Vanda Juranić Lisnić
- Center for Proteomics, Faculty of Medicine, University of Rijeka , Rijeka, Croatia
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zürich , Zürich, Switzerland
| | - Georg Gasteiger
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg , Würzburg, Germany
| | | | - Sonia Tugues
- Institute of Experimental Immunology, University of Zürich , Zürich, Switzerland
- Department of Immunology, Medical Faculty Mannheim, Mannheim Institute for Innate Immunosciences (MI3), Heidelberg University, Heidelberg, Germany
| |
Collapse
|
3
|
Neirinck J, Buysse M, Brdickova N, Perez-Andres M, De Vriendt C, Kerre T, Haerynck F, Bossuyt X, van Dongen JJM, Orfao A, Hofmans M, Bonroy C, Kalina T. The EuroFlow PIDOT external quality assurance scheme: enhancing laboratory performance evaluation in immunophenotyping of rare lymphoid immunodeficiencies. Clin Chem Lab Med 2025; 63:621-635. [PMID: 39423371 DOI: 10.1515/cclm-2024-0749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 09/24/2024] [Indexed: 10/21/2024]
Abstract
OBJECTIVES The development of External Quality Assessment Schemes (EQAS) for clinical flow cytometry (FCM) is challenging in the context of rare (immunological) diseases. Here, we introduce a novel EQAS monitoring the primary immunodeficiency Orientation Tube (PIDOT), developed by EuroFlow, in both a 'wet' and 'dry' format. This EQAS provides feedback on the quality of individual laboratories (i.e., accuracy, reproducibility and result interpretation), while eliminating the need for sample distribution. METHODS In the wet format, marker staining intensities (MedFIs) within landmark cell populations in PIDOT analysis performed on locally collected healthy control (HC) samples, were compared to EQAS targets. In the dry format, participants analyzed centrally distributed PIDOT flow cytometry data (n=10). RESULTS We report the results of six EQAS rounds across 20 laboratories in 11 countries. The wet format (212 HC samples) demonstrated consistent technical performance among laboratories (median %rCV on MedFIs=34.5 %; average failure rate 17.3 %) and showed improvement upon repeated participation. The dry format demonstrated effective proficiency of participants in cell count enumeration (range %rCVs 3.1-7.1 % for the major lymphoid subsets), and in identifying lymphoid abnormalities (79.3 % alignment with reference). CONCLUSIONS The PIDOT-EQAS allows laboratories, adhering to the standardized EuroFlow approach, to monitor interlaboratory variations without the need for sample distribution, and provides them educational support to recognize rare clinically relevant immunophenotypic patterns of primary immunodeficiencies (PID). This EQAS contributes to quality improvement of PID diagnostics and can serve as an example for future flow cytometry EQAS in the context of rare diseases.
Collapse
Affiliation(s)
- Jana Neirinck
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Department of Laboratory Medicine, University Hospital Ghent, Ghent, Belgium
| | - Malicorne Buysse
- Department of Laboratory Medicine, University Hospital Ghent, Ghent, Belgium
| | - Naděžda Brdickova
- CLIP Cytometry, Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Martín Perez-Andres
- Translational and Clinical Research Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca (USAL), Department of Medicine, IBSAL and CIBERONC, University of Salamanca, Salamanca, Spain
- Cancer Research Centre (Instituto de Biologıa Molecular y Celular del Cancer (IBMCC), USAL-CSIC; CIBERONC CB16/12/00400), Institute for Biomedical Research of Salamanca (IBSAL), Department of Medicine and Cytometry Service (NUCLEUS Research Support Platform), University of Salamanca (USAL), Salamanca, Spain
| | - Ciel De Vriendt
- Department of Haematology, University Hospital Ghent, Ghent, Belgium
| | - Tessa Kerre
- Department of Haematology, University Hospital Ghent, Ghent, Belgium
| | - Filomeen Haerynck
- Department of Pediatric Pulmonology and Immunology and PID Research Laboratory, University Hospital Ghent, Ghent, Belgium
| | - Xavier Bossuyt
- Department of Laboratory Medicine, University Hospital Leuven, Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Jacques J M van Dongen
- Translational and Clinical Research Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca (USAL), Department of Medicine, IBSAL and CIBERONC, University of Salamanca, Salamanca, Spain
- Cancer Research Centre (Instituto de Biologıa Molecular y Celular del Cancer (IBMCC), USAL-CSIC; CIBERONC CB16/12/00400), Institute for Biomedical Research of Salamanca (IBSAL), Department of Medicine and Cytometry Service (NUCLEUS Research Support Platform), University of Salamanca (USAL), Salamanca, Spain
| | - Alberto Orfao
- Translational and Clinical Research Program, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca (USAL), Department of Medicine, IBSAL and CIBERONC, University of Salamanca, Salamanca, Spain
- Cancer Research Centre (Instituto de Biologıa Molecular y Celular del Cancer (IBMCC), USAL-CSIC; CIBERONC CB16/12/00400), Institute for Biomedical Research of Salamanca (IBSAL), Department of Medicine and Cytometry Service (NUCLEUS Research Support Platform), University of Salamanca (USAL), Salamanca, Spain
| | - Mattias Hofmans
- Department of Laboratory Medicine, University Hospital Ghent, Ghent, Belgium
| | - Carolien Bonroy
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Department of Laboratory Medicine, University Hospital Ghent, Ghent, Belgium
| | - Tomas Kalina
- CLIP Cytometry, Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| |
Collapse
|
4
|
Bordoni V, Cinicola BL, Piano Mortari E, Castilletti C, Guarracino F, Albano C, Accordini S, Baban A, Di Sabatino A, Rossi CM, Lenti MV, Zicari AM, Cirelli R, Spada M, Forni GL, Quinti I, Algeri M, Casale M, Perrotta S, Locatelli F, Agrati C, Carsetti R. Impairment of Innate Immunity and Depletion of Vaccine-Induced Memory B and T Cells in the Absence of the Spleen. Am J Hematol 2025. [PMID: 39953916 DOI: 10.1002/ajh.27634] [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: 02/01/2025] [Accepted: 02/03/2025] [Indexed: 02/17/2025]
Abstract
Splenectomy or congenital asplenia is associated with severe reduction of memory B cells and increased risk of fulminant sepsis by encapsulated bacteria. Current guidelines recommend vaccinations against these pathogens before or after splenectomy, but the longevity of immunity acquired after splenectomy has not been determined. The impact of splenectomy on innate immune cells is unknown. We analyzed frequency, differentiation stage, and function of innate and adaptive immunity in the peripheral blood of adult (n = 41) and pediatric (n = 14) patients splenectomized or born asplenic and in spleens of solid organ donors. The absence of the spleen impacts the B-cell compartment, causing a significant increase of circulating immature transitional and depletion of memory B cells. Using SARS-CoV-2 vaccination as a model, we show that 1 year after the last immunization, despite normal levels of neutralizing antibodies, memory B and T cells were significantly reduced. Analysis of post-pandemic spleens shows that spike-specific memory B and T cells homed to the spleen. We also show a previously unrecognized role of the spleen in the homeostasis of innate NK and Vδ2 T cells. These populations showed altered phenotype and impaired function in the adults, but not in children, suggesting that other tissues may support innate cell development during early life. The reduced function of innate lymphocytes must be considered as an additional immune impairment and risk factor. These findings emphasize the spleen's irreplaceable role in maintaining immune memory across all ages and suggest that its absence contributes to dysfunctions of innate and adaptive immunity in adults.
Collapse
Affiliation(s)
- Veronica Bordoni
- Department of Hematology/Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, Rome, Italy
| | - Bianca Laura Cinicola
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- Pediatric Unit, NESMOS Department, Sant'Andrea University Hospital, Sapienza University of Rome, Rome, Italy
| | - Eva Piano Mortari
- Research Area of Immunology, B-Cell Lab, Bambino Gesù Children's Hospital, Rome, Italy
| | - Concetta Castilletti
- Department of Infectious-Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital, Verona, Italy
| | - Federica Guarracino
- Department of Hematology/Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, Rome, Italy
| | - Christian Albano
- Research Area of Immunology, B-Cell Lab, Bambino Gesù Children's Hospital, Rome, Italy
| | - Silvia Accordini
- Department of Infectious-Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital, Verona, Italy
| | - Anwar Baban
- Cardiogenetic Centre, Rare Diseases and Medical Genetics Units, Bambino Gesù Children's Hospital, Rome, Italy
| | - Antonio Di Sabatino
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy
- First Department of Internal Medicine, San Matteo Hospital Foundation, Pavia, Italy
| | - Carlo Maria Rossi
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy
- First Department of Internal Medicine, San Matteo Hospital Foundation, Pavia, Italy
| | - Marco Vincenzo Lenti
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy
- First Department of Internal Medicine, San Matteo Hospital Foundation, Pavia, Italy
| | - Anna Maria Zicari
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Riccardo Cirelli
- Unit of Hepato-Biliary-Pancreatic Surgery, Liver and Kidney Transplantation, Bambino Gesù Children's Hospital, Rome, Italy
| | - Marco Spada
- Unit of Hepato-Biliary-Pancreatic Surgery, Liver and Kidney Transplantation, Bambino Gesù Children's Hospital, Rome, Italy
| | - Gian Luca Forni
- Unit of Hematology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
- ForAnemia Foundation, Genoa, Italy
| | - Isabella Quinti
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Mattia Algeri
- Department of Hematology/Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, Rome, Italy
- Department of Health Sciences, Magna Graecia University, Catanzaro, Italy
| | - Maddalena Casale
- Department of Woman, Child and General and Specialized Surgery, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Silverio Perrotta
- Department of Woman, Child and General and Specialized Surgery, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Franco Locatelli
- Department of Hematology/Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, Rome, Italy
- Catholic University of the Sacred Heart, Rome, Italy
| | - Chiara Agrati
- Department of Hematology/Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, Rome, Italy
| | - Rita Carsetti
- Research Area of Immunology, B-Cell Lab, Bambino Gesù Children's Hospital, Rome, Italy
| |
Collapse
|
5
|
Joosse HJ, Chumsaeng-Reijers C, Huisman A, Hoefer IE, van Solinge WW, Haitjema S, van Es B. Haematology dimension reduction, a large scale application to regular care haematology data. BMC Med Inform Decis Mak 2025; 25:75. [PMID: 39939843 PMCID: PMC11823074 DOI: 10.1186/s12911-025-02899-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 01/28/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND The routine diagnostic process increasingly entails the processing of high-volume and high-dimensional data that cannot be directly visualised. This processing may provide scaling issues that limit the implementation of these types of data into research as well as integrated diagnostics in routine care. Here, we investigate whether we can use existing dimension reduction techniques to provide visualisations and analyses for a complete bloodcount (CBC) while maintaining representativeness of the original data. We considered over 3 million CBC measurements encompassing over 70 parameters of cell frequency, size and complexity from the UMC Utrecht UPOD database. We evaluated PCA as an example of a linear dimension reduction techniques and UMAP, TriMap and PaCMAP as non-linear dimension reduction techniques. We assessed their technical performance using quality metrics for dimension reduction as well as biological representation by evaluating preservation of diurnal, age and sex patterns, cluster preservation and the identification of leukemia patients. RESULTS We found that, for clinical hematology data, PCA performs systematically better than UMAP, TriMap and PaCMAP in representing the underlying data. Biological relevance was retained for periodicity in the data. However, we also observed a decrease in predictive performance of the reduced data for both age and sex, as well as an overestimation of clusters within the reduced data. Finally, we were able to identify the diverging patterns for leukemia patients after use of dimensionality reduction methods. CONCLUSIONS We conclude that for hematology data, the use of unsupervised dimension reduction techniques should be limited to data visualization applications, as implementing them in diagnostic pipelines may lead to decreased quality of integrated diagnostics in routine care.
Collapse
Affiliation(s)
- Huibert-Jan Joosse
- Central Diagnostic Laboratory, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, 3584 CX, The Netherlands
| | - Chontira Chumsaeng-Reijers
- Central Diagnostic Laboratory, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, 3584 CX, The Netherlands
| | - Albert Huisman
- Central Diagnostic Laboratory, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, 3584 CX, The Netherlands
| | - Imo E Hoefer
- Central Diagnostic Laboratory, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, 3584 CX, The Netherlands
| | - Wouter W van Solinge
- Central Diagnostic Laboratory, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, 3584 CX, The Netherlands
| | - Saskia Haitjema
- Central Diagnostic Laboratory, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, 3584 CX, The Netherlands
| | - Bram van Es
- Central Diagnostic Laboratory, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, 3584 CX, The Netherlands.
| |
Collapse
|
6
|
Chen J, Ionita M, Feng Y, Lu Y, Orzechowski P, Garai S, Hassinger K, Bao J, Wen J, Duong-Tran D, Wagenaar J, McKeague ML, Painter MM, Mathew D, Pattekar A, Meyer NJ, Wherry EJ, Greenplate AR, Shen L. Automated cytometric gating with human-level performance using bivariate segmentation. Nat Commun 2025; 16:1576. [PMID: 39939580 PMCID: PMC11821879 DOI: 10.1038/s41467-025-56622-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 01/21/2025] [Indexed: 02/14/2025] Open
Abstract
Recent advances in cytometry have enabled high-throughput data collection with multiple single-cell protein expression measurements. The significant biological and technical variance in cytometry has posed a formidable challenge during the gating process, especially for the initial pre-gates which deal with unpredictable events, such as debris and technical artifacts. To mitigate the labor-intensive manual gating process, we propose UNITO, a framework to rigorously identify the hierarchical cytometric subpopulations. UNITO transforms a cell-level classification task into an image-based segmentation problem. The framework is validated on three independent cohorts (two mass cytometry and one flow cytometry datasets). We compare its results with previous automated methods using the consensus of at least four experienced immunologists. UNITO outperforms existing methods and deviates from human consensus by no more than any individual does. UNITO can reproduce a similar contour compared to manual gating for post-hoc inspection, and it also allows parallelization of samples for faster processing.
Collapse
Affiliation(s)
- Jiong Chen
- Department of Bioengineering, University of Pennsylvania School of Engineering and Applied Science, Philadelphia, PA, USA
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Matei Ionita
- Department of Systems Pharmacology & Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Immunology and Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Yanbo Feng
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Yinfeng Lu
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Mathematics, University of Pennsylvania School of Arts and Sciences, Philadelphia, PA, USA
| | - Patryk Orzechowski
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Automatics and Robotics, AGH University of Science and Technology, Krakow, Poland
| | - Sumita Garai
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kenneth Hassinger
- Department of Systems Pharmacology & Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jingxuan Bao
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Junhao Wen
- Laboratory of AI and Biomedical Science (LABS), University of Southern California, Los Angeles, CA, USA
| | - Duy Duong-Tran
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Mathematics, United States Naval Academy, Annapolis, MD, USA
| | - Joost Wagenaar
- Department of Systems Pharmacology & Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Michelle L McKeague
- Department of Systems Pharmacology & Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Immunology and Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Mark M Painter
- Department of Systems Pharmacology & Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Immunology and Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Divij Mathew
- Department of Systems Pharmacology & Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Immunology and Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ajinkya Pattekar
- Department of Systems Pharmacology & Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Immunology and Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Nuala J Meyer
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - E John Wherry
- Department of Systems Pharmacology & Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Immunology and Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Allison R Greenplate
- Department of Systems Pharmacology & Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Immunology and Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
7
|
Morse MA, Crosby EJ, Halperin DM, Uronis HE, Hsu SD, Hurwitz HI, Rushing C, Bolch EK, Warren DA, Moyer AN, Lowe ME, Niedzwiecki D. Phase Ib/II study of Pembrolizumab with Lanreotide depot for advanced, progressive Gastroenteropancreatic neuroendocrine tumors (PLANET). J Neuroendocrinol 2025:e13496. [PMID: 39933708 DOI: 10.1111/jne.13496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 11/28/2024] [Accepted: 01/25/2025] [Indexed: 02/13/2025]
Abstract
While performing a study of immune checkpoint blockade with the anti-PD-1 antibody pembrolizumab combined with the somatostatin analogue (SSA) lanreotide in patients with low- and intermediate-grade gastroenteropancreatic neuroendocrine tumors (GEP-NETs), we studied whether there were any immune correlates of response to the anti-PD-1 therapy that could guide future attempts to integrate immunotherapy into the treatment of NETs. Patients with grade 1 and 2 GEP-NETs who had progressed on a prior SSA received lanreotide 90 mg subcutaneously and pembrolizumab 200 mg intravenously every 3 weeks until progression or intolerable toxicity. Objective response rate (ORR) at any time in the study, clinical benefit rate (CBR, defined as stable disease or better), progression-free survival (PFS), and overall survival (OS) were measured. Changes in T cell subsets in peripheral blood before and during therapy were analyzed by multiparameter mass cytometry (CyTOF). Archived tissue samples were analyzed for PD-L1 expression and TIL infiltration. Twenty-two (22) patients (GI/pancreatic 14/8, median Ki67 7% [IQR 4, 10%], median 1.5 prior systemic therapies [range 1-4]) were enrolled. Among the GI-NETs, there was one partial response, the CBR was 50%, the median PFS was 8.5 months, and the median OS was 32.7 months. No responses were seen in pancreatic NETs, which had 0% CBR, a PFS of 2.7 months, and an OS of 23.9 months. Of the 16 analyzable tumors, 6 had detectable PD-L1 expression and 15 had detectable TILs. Neither TILs nor PD-L1 expression correlated with ORR or CBR. However, clinical benefit (SD or better) was associated with peripheral blood on-treatment effector memory T cell activation and progressive disease was associated with baseline peripheral blood regulatory T cell (Treg) activation. We conclude that immune checkpoint blockade had low activity in unselected patients with grade 1 and 2 GEP-NETs. Further study of strategies to reduce Treg activation or enhance effector memory activation during immunotherapy is warranted.
Collapse
Affiliation(s)
- Michael A Morse
- Division of Medical Oncology, Duke University Department of Medicine, Durham, North Carolina, USA
| | - Erika J Crosby
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Daniel M Halperin
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hope E Uronis
- Division of Medical Oncology, Duke University Department of Medicine, Durham, North Carolina, USA
| | - S David Hsu
- Division of Medical Oncology, Duke University Department of Medicine, Durham, North Carolina, USA
| | - Herbert I Hurwitz
- Division of Medical Oncology, Duke University Department of Medicine, Durham, North Carolina, USA
| | - Christel Rushing
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Emily K Bolch
- Duke Cancer Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Dana A Warren
- Duke Cancer Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Ashley N Moyer
- Duke Cancer Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Melissa E Lowe
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Donna Niedzwiecki
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
8
|
Riemann L, Gutierrez R, Odak I, Barros-Martins J, Roesner LM, Leon Lara X, Falk C, Schulz TF, Hansen G, Werfel T, Förster R. Integrative deep immune profiling of the elderly reveals systems-level signatures of aging, sex, smoking, and clinical traits. EBioMedicine 2025; 112:105558. [PMID: 39862806 DOI: 10.1016/j.ebiom.2025.105558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/13/2024] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Aging increases disease susceptibility and reduces vaccine responsiveness, highlighting the need to better understand the aging immune system and its clinical associations. Studying the human immune system, however, remains challenging due to its complexity and significant inter-individual variability. METHODS We conducted an immune profiling study of 550 elderly participants (≥60 years) and 100 young controls (20-40 years) from the RESIST Senior Individuals (SI) cohort. Extensive demographic, clinical, and laboratory data were collected. Multi-color spectral flow cytometry and 48-plex plasma cytokine assays were used for deep immune phenotyping. Data were analyzed using unsupervised clustering and multi-dataset integration approaches. FINDINGS We studied 97 innate and adaptive immune cell populations, revealing intricate age- and sex-related changes in the elderly immune system. Our large sample size allowed detection of even subtle changes in cytokines and immune cell clusters. Integrative analysis combining clinical, laboratory, and immunological data revealed systems-level aging signatures, including shifts in specific immune cell subpopulations and cytokine concentrations (e.g., HGF and CCL27). Additionally, we identified unique immune signatures associated with smoking, obesity, and diseases such as osteoporosis, heart failure, and gout. INTERPRETATION This study provides one of the most comprehensive immune profiles of elderly individuals, uncovering high-resolution immune changes associated with aging. Our findings highlight clinically relevant immune signatures that enhance our understanding of aging-related diseases and could guide future research into new treatments, offering translational insights into human health and aging. FUNDING Deutsche Forschungsgemeinschaft (DFG, German Research Foundation) under Germany's Excellence Strategy-EXC 2155-project number 390874280.
Collapse
Affiliation(s)
- Lennart Riemann
- Institute of Immunology, Hannover Medical School, Hannover, Germany; Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany.
| | | | - Ivan Odak
- Institute of Immunology, Hannover Medical School, Hannover, Germany; The Tisch Cancer Institute, Icahn School of Medicine, New York, USA
| | - Joana Barros-Martins
- Institute of Immunology, Hannover Medical School, Hannover, Germany; Department of Microbiology and Immunology, Columbia University Medical Center, New York, USA
| | - Lennart M Roesner
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Ximena Leon Lara
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Christine Falk
- Institute of Transplantation Immunology, Hannover Medical School, Hannover, Germany; German Centre for Infection Research, Partner Site Hannover-Braunschweig, Hannover, Germany
| | - Thomas F Schulz
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany; German Centre for Infection Research, Partner Site Hannover-Braunschweig, Hannover, Germany; Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Gesine Hansen
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany; German Center of Lung Research (DZL), BREATH, Hannover, Germany
| | - Thomas Werfel
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany; German Centre for Infection Research, Partner Site Hannover-Braunschweig, Hannover, Germany.
| |
Collapse
|
9
|
Spies NC, Rangel A, English P, Morrison M, O’Fallon B, Ng DP. Machine Learning Methods in Clinical Flow Cytometry. Cancers (Basel) 2025; 17:483. [PMID: 39941850 PMCID: PMC11816335 DOI: 10.3390/cancers17030483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/27/2025] [Accepted: 01/28/2025] [Indexed: 02/16/2025] Open
Abstract
This review will explore the integration of machine learning (ML) techniques to enhance the analysis of increasingly complex and voluminous flow cytometry data, as traditional manual methods are insufficient for handling this data. We attempt to provide a comprehensive introduction to ML in flow cytometry, detailing the transition from manual gating to computational methods and emphasizing the importance of data quality. Key ML techniques are discussed, including supervised learning methods like logistic regression, support vector machines, and neural networks, which rely on labeled data to classify disease states. Unsupervised methods, such as k-means clustering, FlowSOM, UMAP, and t-SNE, are highlighted for their ability to identify novel cell populations without predefined labels. We also delve into newer semi-supervised and weakly supervised methods, which leverage partial labeling to improve model performance. Practical aspects of implementing ML in clinical settings are addressed, including regulatory considerations, data preprocessing, model training, validation, and the importance of generalizability, and we underscore the collaborative effort required among pathologists, data scientists, and laboratory professionals to ensure robust model development and deployment. Finally, we show the transformative potential of ML in flow cytometry in uncovering new biological insights through advanced computational techniques.
Collapse
Affiliation(s)
- Nicholas C. Spies
- Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
- ARUP Laboratories, Division of Applied Artificial Intelligence, Institute for Research and Innovation, Salt Lake City, UT 84108, USAbrendan.o’
| | - Alexandra Rangel
- ARUP Laboratories, Division of Applied Artificial Intelligence, Institute for Research and Innovation, Salt Lake City, UT 84108, USAbrendan.o’
| | - Paul English
- ARUP Laboratories, Division of Applied Artificial Intelligence, Institute for Research and Innovation, Salt Lake City, UT 84108, USAbrendan.o’
| | - Muir Morrison
- ARUP Laboratories, Division of Applied Artificial Intelligence, Institute for Research and Innovation, Salt Lake City, UT 84108, USAbrendan.o’
| | - Brendan O’Fallon
- ARUP Laboratories, Division of Applied Artificial Intelligence, Institute for Research and Innovation, Salt Lake City, UT 84108, USAbrendan.o’
| | - David P. Ng
- Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
- ARUP Laboratories, Division of Applied Artificial Intelligence, Institute for Research and Innovation, Salt Lake City, UT 84108, USAbrendan.o’
| |
Collapse
|
10
|
Gao Y, Liu R, Shi J, Shan W, Zhou H, Chen Z, Yue X, Zhang J, Luo Y, Pan W, Zhao X, Zeng X, Yin W, Xiao H. Clonal GZMK +CD8 + T cells are identified as a hallmark of the pathogenesis of cGVHD-induced bronchiolitis obliterans syndrome after allogeneic hematopoietic stem cell transplantation. EBioMedicine 2025; 112:105535. [PMID: 39740295 PMCID: PMC11750515 DOI: 10.1016/j.ebiom.2024.105535] [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: 04/09/2024] [Revised: 12/13/2024] [Accepted: 12/17/2024] [Indexed: 01/02/2025] Open
Abstract
BACKGROUND Bronchiolitis obliterans syndrome (BOS) is one of the most devastating outcomes of chronic graft-versus-host disease (cGVHD) after allogeneic hematopoietic stem cell transplantation (allo-HSCT). This remains an area of unmet clinical need for optimal therapy for BOS patients partly due to the limited understanding of pathogenic mechanisms. METHODS We collected blood samples from 22 patients with cGVHD and 11 patients without cGVHD following allo-HSCT. By applying a combination of mass cytometry (CyTOF), RNA-sequencing and the quantitative cytokine array, we discovered a new cellular hallmarker of patients with cGVHD-BOS. This finding was further validated in cGVHD-BOS murine models by using single-cell RNA sequencing (scRNA-seq) and paired single-cell V(D)J sequencing analyses. FINDINGS We revealed that circulating Granzyme K (GZMK)-expressing CD8+ T cells with increased expression of CCR5 were accumulated in cGVHD-BOS patients, and GZMK can induce the expression of fibrosis-essential proteins, collagen type I alpha 1 chain (COL1A1) and fibronectin (FN1), in human fibroblasts. As compared to those of control mice, GZMK+CD8+ T cells in the lungs of cGVHD-BOS mice were undergoing significant infiltration and clonal hyperexpansion, with more cytotoxic, pro-inflammatory, migratory and exhausted phenotypes. Moreover, we screened small-molecule drugs and revealed that Bosutinib, the second-generation BCR-ABL1-targeting tyrosine kinase inhibitor (TKI), could inhibit GZMK expression in CD8+ T cells and reduce lung stiffness and pulmonary fibrosis in cGVHD-BOS mice. INTERPRETATION This study provides proof-of-principle evidence for clonal GZMK+CD8+ T cells as an unexplored contributor to the pathogenesis of cGVHD-BOS, which can be an underlying biomarker for treatment. FUNDING This work was supported by the National Natural Science Foundation of China (No. 82170141, 82100123, 81870136), and "Pioneer" and "Leading Goose" R&D Program of Zhejiang (grant No. 2022C03012).
Collapse
Affiliation(s)
- Yang Gao
- Department of Hematology and Cell Therapy, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang province, PR China
| | - Ruixiang Liu
- Zhejiang Puluoting Health Technology Co., Ltd, Hangzhou, Zhejiang province, PR China
| | - Jiawei Shi
- Key Laboratory for Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, Zhejiang province, PR China
| | - Wei Shan
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang province, PR China
| | - Hongyu Zhou
- Department of Hematology and Cell Therapy, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang province, PR China
| | - Zhi Chen
- Department of Hematology and Cell Therapy, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang province, PR China
| | - Xiaoyan Yue
- Department of Hematology and Cell Therapy, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang province, PR China
| | - Jie Zhang
- Department of Hematology and Cell Therapy, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang province, PR China
| | - Yi Luo
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang province, PR China
| | - Wenjue Pan
- Department of Hematology and Cell Therapy, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang province, PR China
| | - Xiujie Zhao
- Department of Hematology and Cell Therapy, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang province, PR China
| | - Xun Zeng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang province, PR China.
| | - Weiwei Yin
- Key Laboratory for Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, Zhejiang province, PR China; Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, College of Biomedical Engineering and Instrument of Science, Zhejiang University, Hangzhou, Zhejiang province, PR China.
| | - Haowen Xiao
- Department of Hematology and Cell Therapy, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang province, PR China.
| |
Collapse
|
11
|
Tandaric L, Auranen A, Kleinmanns K, DePont Christensen R, Vestrheim Thomsen LC, Wogsland CE, McCormack E, Mäenpää J, Madsen K, Stampe Petersson K, Mirza MR, Bjørge L. Peripheral blood leukocyte signatures as biomarkers in relapsed ovarian cancer patients receiving combined anti-CD73/anti-PD-L1 immunotherapy in arm A of the NSGO-OV-UMB1/ENGOT-OV30 trial. Mol Oncol 2025. [PMID: 39887612 DOI: 10.1002/1878-0261.13811] [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: 01/02/2025] [Accepted: 01/20/2025] [Indexed: 02/01/2025] Open
Abstract
Immune checkpoint inhibitors have demonstrated limited efficacy in overcoming immunosuppression in patients with epithelial ovarian cancer (EOC). Although certain patients experience long-term treatment benefit, reliable biomarkers for responder pre-selection and the distinction of dominant immunosuppressive mechanisms have yet to be identified. Here, we used a 40-marker suspension mass cytometry panel to comprehensively phenotype peripheral blood leukocytes sampled over time from patients with relapsed EOC who underwent combination oleclumab (anti-CD73) and durvalumab (anti-PD-L1) immunotherapy in the NSGO-OV-UMB1/ENGOT-OV30 trial. We found that survival duration was impacted by baseline abundances of total peripheral blood mononuclear cells. Longitudinal analyses revealed a significant increase in CD14+CD16- myeloid cells during treatment, with significant expansion of monocytic myeloid-derived suppressor cells occurring in patients with shorter progression-free survival, who additionally showed a continuous decrease in central memory T-cell abundances. All patients demonstrated significant PD-L1 upregulation over time on most T-cell subsets. Higher CD73 and IDO1 expression on certain leukocytes at baseline significantly positively correlated with longer progression-free survival. Overall, our study proposes potential biomarkers for EOC immunotherapy personalization and response monitoring; however, further validation in larger studies is needed.
Collapse
Affiliation(s)
- Luka Tandaric
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Norway
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
| | - Annika Auranen
- Department of Obstetrics and Gynecology and Tays Cancer Centre, Tampere University Hospital, Finland
- Nordic Society of Gynaecological Oncology - Clinical Trial Unit (NSGO-CTU), Tampere, Finland
| | - Katrin Kleinmanns
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Norway
| | - René DePont Christensen
- Nordic Society of Gynaecological Oncology - Clinical Trial Unit (NSGO-CTU), Copenhagen, Denmark
| | - Liv Cecilie Vestrheim Thomsen
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Norway
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
- Department of Health Registry Research and Development, Norwegian Institute of Public Health, Oslo, Norway
| | - Cara Ellen Wogsland
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Norway
- Kinn Therapeutics AS, Bergen, Norway
| | - Emmet McCormack
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Norway
- Centre for Pharmacy, Department of Clinical Science, University of Bergen, Norway
- Department of Internal Medicine, Hematology Section, Haukeland University Hospital, Bergen, Norway
| | - Johanna Mäenpää
- Department of Obstetrics and Gynecology and Tays Cancer Centre, Tampere University Hospital, Finland
- Nordic Society of Gynaecological Oncology - Clinical Trial Unit (NSGO-CTU), Tampere, Finland
- Faculty of Medicine and Health Technology, Tampere University, Finland
| | - Kristine Madsen
- Nordic Society of Gynaecological Oncology - Clinical Trial Unit (NSGO-CTU), Copenhagen, Denmark
| | - Karen Stampe Petersson
- Nordic Society of Gynaecological Oncology - Clinical Trial Unit (NSGO-CTU), Copenhagen, Denmark
| | - Mansoor Raza Mirza
- Nordic Society of Gynaecological Oncology - Clinical Trial Unit (NSGO-CTU), Copenhagen, Denmark
- Department of Oncology, Rigshospitalet, Copenhagen University Hospital, Denmark
| | - Line Bjørge
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Norway
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
12
|
Chan L, Pinedo K, Stabile MA, Hamlin RE, Pienkos SM, Ratnasiri K, Yang S, Blomkalns AL, Nadeau KC, Pulendran B, O'Hara R, Rogers AJ, Holmes SP, Blish CA. Prior vaccination prevents overactivation of innate immune responses during COVID-19 breakthrough infection. Sci Transl Med 2025; 17:eadq1086. [PMID: 39879318 DOI: 10.1126/scitranslmed.adq1086] [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: 05/03/2024] [Revised: 09/10/2024] [Accepted: 12/16/2024] [Indexed: 01/31/2025]
Abstract
At this stage in the COVID-19 pandemic, most infections are "breakthrough" infections that occur in individuals with prior severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) exposure. To refine long-term vaccine strategies against emerging variants, we examined both innate and adaptive immunity in breakthrough infections. We performed single-cell transcriptomic, proteomic, and functional profiling of primary and breakthrough infections to compare immune responses from unvaccinated and vaccinated individuals during the SARS-CoV-2 Delta wave. Breakthrough infections were characterized by a less activated transcriptomic profile in monocytes and natural killer cells, with induction of pathways limiting monocyte migratory potential and natural killer cell proliferation. Furthermore, we observed a female-specific increase in transcriptomic and proteomic activation of multiple innate immune cell subsets during breakthrough infections. These insights suggest that prior SARS-CoV-2 vaccination prevents overactivation of innate immune responses during breakthrough infections with discernible sex-specific patterns and underscore the potential of harnessing vaccines in mitigating pathologic immune responses resulting from overactivation.
Collapse
Affiliation(s)
- Leslie Chan
- Stanford Immunology Program, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kassandra Pinedo
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mikayla A Stabile
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Rebecca E Hamlin
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Shaun M Pienkos
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kalani Ratnasiri
- Stanford Immunology Program, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Samuel Yang
- Department of Emergency Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Andra L Blomkalns
- Department of Emergency Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kari C Nadeau
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ruth O'Hara
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Angela J Rogers
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Susan P Holmes
- Department of Statistics, Stanford University, Stanford, CA 94305, USA
| | - Catherine A Blish
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Stanford Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA 94305, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| |
Collapse
|
13
|
Omdahl KI, Bermea RS, Fleming R, Kimler K, Kaminski J, Hariri LP, Ly A, Rui X, Cagnin L, Lane J, Gerdemann U, Blazar BR, Tkachev V, Kean LS. Organ-specific microenvironments drive divergent T cell evolution in acute graft-versus-host disease. Sci Transl Med 2025; 17:eads1298. [PMID: 39879321 DOI: 10.1126/scitranslmed.ads1298] [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: 07/30/2024] [Accepted: 10/28/2024] [Indexed: 01/31/2025]
Abstract
Tissue-specific T cell immune responses play a critical role in maintaining organ health but can also drive immune pathology during both autoimmunity and alloimmunity. The mechanisms controlling intratissue T cell programming remain unclear. Here, we leveraged a nonhuman primate model of acute graft-versus-host disease (aGVHD) after allogeneic hematopoietic stem cell transplantation to probe the biological underpinnings of tissue-specific alloimmune disease using a comprehensive systems immunology approach including multiparameter flow cytometry, population-based transcriptional profiling, and multiplexed single-cell RNA sequencing and TCR sequencing. Transcriptional profiling revealed substantial biological differences between T cells infiltrating the lung and liver during aGVHD. These included enrichment for transcriptional pathways controlling extracellular matrix remodeling and chemotaxis in the lung and enrichment for transcriptional pathways linked to nucleic acid metabolism and proliferation in the liver. Single-cell RNA sequencing and TCR sequencing substantiated divergent organ-specific transcriptional programing of tissue-infiltrating T cells, which was linked to clonal expansion, with expanded clones progressively enriched for C-X3-C motif chemokine receptor 1 (CX3CR1)-expressing CD8 effector T cells in the lung and eomesodermin (EOMES)-expressing CD8 effector-memory T cells in the liver. This divergent evolution of T cells was maintained even for T cells sharing the same TCRs, indicating its independence from antigen specificity. Together, these results provide insights into the role that tissue microenvironment-derived signals play in local T cell transcriptional programming during alloimmune-mediated clonal expansion and suggest potential opportunities to develop tissue-specific therapeutics to curtail pathogenic immunity after transplant.
Collapse
Affiliation(s)
- Kayleigh Ingersoll Omdahl
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Rene S Bermea
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
- Lung Transplant Program, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Center for Transplantation Sciences, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Ryan Fleming
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Kyle Kimler
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
- Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - James Kaminski
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
- Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Lida P Hariri
- Harvard Medical School, Boston, MA 02115, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Amy Ly
- Harvard Medical School, Boston, MA 02115, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Xianliang Rui
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Lorenzo Cagnin
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Jennifer Lane
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Ulrike Gerdemann
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Bruce R Blazar
- Division of Pediatric Blood and Marrow Transplantation & Cellular Therapy, Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Victor Tkachev
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
- Center for Transplantation Sciences, Massachusetts General Hospital, Boston, MA 02129, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Leslie S Kean
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| |
Collapse
|
14
|
Zhu A, Chen Z, Yan Q, Jiang M, Liu X, Li Z, Li N, Tang C, Jian W, He J, Chen L, Cheng J, Chen C, Tang T, Xu Z, Hu Q, Li F, Wang Y, Sun J, Zhuang Z, Wen L, Zhuo J, Liu D, Zhang Y, Huang X, Li S, Zeng Q, Chen F, Zhou L, Liu D, Zhong C, Chen Y, Li S, Liang K, Zhong N, Zhang X, Chen J, Chen X, Xu Y, Zhong N, Zhao J, Zhao J. Robust mucosal SARS-CoV-2-specific T cells effectively combat COVID-19 and establish polyfunctional resident memory in patient lungs. Nat Immunol 2025:10.1038/s41590-024-02072-9. [PMID: 39875584 DOI: 10.1038/s41590-024-02072-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 12/17/2024] [Indexed: 01/30/2025]
Abstract
Mucosal antigen-specific T cells are pivotal for pathogen clearance and immune modulation in respiratory infections. Dysregulated T cell responses exacerbate coronavirus disease 2019 severity, marked by cytokine storms and respiratory failure. Despite extensive description in peripheral blood, the characteristics of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific T cells in the lungs remain elusive. Here we conducted integrated single-cell profiling of SARS-CoV-2-specific T cells in 122 bronchoalveolar lavage fluid (BALF) and 280 blood samples from 159 patients, including 27 paired BALF and blood samples from 24 patients. SARS-CoV-2-specific T cells were robustly elicited in BALF irrespective of prior vaccination, correlating with diminished viral loads, lessened systemic inflammation and improved respiratory function. SARS-CoV-2-specific T cells in BALF exhibited profound activation, along with proliferative and multi-cytokine-producing capabilities and a glycolysis-driven metabolic signature, which were distinct from those observed in peripheral blood mononuclear cells. After viral clearance, these specific T cells maintained a polyfunctional tissue-resident memory phenotype, highlighting their critical roles in infection control and long-term protection.
Collapse
Affiliation(s)
- Airu Zhu
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhao Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qihong Yan
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mei Jiang
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xuesong Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Critical Care Medicine, State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhengtu Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Na Li
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, China
| | - Chunli Tang
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenhua Jian
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | | | - Lan Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou, China
| | - Jinling Cheng
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Canjie Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Tian Tang
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhiwei Xu
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qingtao Hu
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China
| | - Fang Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yanqun Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jing Sun
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhen Zhuang
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Liyan Wen
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jianfen Zhuo
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Donglan Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yanjun Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaofang Huang
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Suxiang Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qiuhui Zeng
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Fangli Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Liang Zhou
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Critical Care Medicine, State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Dongdong Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Critical Care Medicine, State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Changhao Zhong
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yu Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shiyue Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Kangli Liang
- Guangdong Hospital of Integrated Traditional Chinese and Western Medicine, Foshan, China
| | - Na Zhong
- Shenzhen Peacock Biotechnology Co. Ltd, Shenzhen, China
| | - Xinmei Zhang
- Shenzhen Peacock Biotechnology Co. Ltd, Shenzhen, China
| | - Jiekai Chen
- Center for Cell Lineage and Development, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xiaobo Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Yonghao Xu
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Department of Critical Care Medicine, State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Guangzhou National Laboratory, Guangzhou, China.
| | - Nanshan Zhong
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Guangzhou National Laboratory, Guangzhou, China.
| | - Jingxian Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Guangzhou National Laboratory, Guangzhou, China.
| | - Jincun Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Guangzhou National Laboratory, Guangzhou, China.
- Shanghai Institute for Advanced Immunochemical Studies, School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
- National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
15
|
Quintelier KLA, Willemsen M, Bosteels V, Aerts JGJV, Saeys Y, Van Gassen S. CytoNorm 2.0: A flexible normalization framework for cytometry data without requiring dedicated controls. Cytometry A 2025. [PMID: 39871681 DOI: 10.1002/cyto.a.24910] [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: 10/30/2023] [Revised: 11/07/2024] [Accepted: 11/17/2024] [Indexed: 01/29/2025]
Abstract
Cytometry is a single cell, high-dimensional, high-throughput technique that is being applied across a range of disciplines. However, many elements alongside the data acquisition process might give rise to technical variation in the dataset, called batch effects. CytoNorm is a normalization algorithm for batch effect removal in cytometry data that was originally published in 2020 and has been applied on a variety of datasets since then. Here, we present CytoNorm 2.0, discussing new, illustrative use cases to increase the applicability of the algorithm and showcasing new visualizations that enable thorough quality control and understanding of the normalization process. We explain how CytoNorm can be used without the need for technical replicates or controls, show how the goal distribution can be tailored toward the experimental design and we elaborate on the choice of markers for CytoNorm's internal FlowSOM clustering step.
Collapse
Affiliation(s)
- Katrien L A Quintelier
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
- Data Mining and Modeling for Biomedicine Group, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, The Netherlands
| | - Marcella Willemsen
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, The Netherlands
| | - Victor Bosteels
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Laboratory for ER Stress and Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Joachim G J V Aerts
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, The Netherlands
| | - Yvan Saeys
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
- Data Mining and Modeling for Biomedicine Group, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Sofie Van Gassen
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
- Data Mining and Modeling for Biomedicine Group, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| |
Collapse
|
16
|
Rodríguez IJ, Parra-López CA. Markers of immunosenescence in CMV seropositive healthy elderly adults. FRONTIERS IN AGING 2025; 5:1436346. [PMID: 39916725 PMCID: PMC11798936 DOI: 10.3389/fragi.2024.1436346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 12/31/2024] [Indexed: 02/09/2025]
Abstract
A significant increase in life expectancy has accompanied the growth of the world's population. Approximately 10% of the global population are adults over 60, and it is estimated that 2050 this figure will double. This increase in the proportion of older adults leads to a more significant burden of age-related diseases. Immunosenescence predisposes elderly individuals to a higher incidence of infectious and chronic non-communicable diseases with higher mortality rates. Despite advances in research, it is necessary to evaluate the cellular characteristics of the aging immune system in populations with a high incidence of latent viruses such as cytomegalovirus (CMV). In this sense, this work aimed to identify senescence markers in cells of the innate and adaptive immune system in healthy older adults with CMV infection. We observed that older adults present an increase in the population of CD14+CD16+ intermediate monocytes, an expansion of CD56neg NK cells with an increase in the expression of CD57, as well as a decrease in the naïve CD4+ and CD8+ T cells, accompanied by an increased expression of senescence markers CD57 and KLRG1 in effector CD8+ T cells.
Collapse
Affiliation(s)
- Ivón Johanna Rodríguez
- Grupo de profundización en Kinesioterapia, Departamento de Movimiento Corporal Humano, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Carlos Alberto Parra-López
- Grupo de Inmunología y Medicina Traslacional, Departamento de Microbiología, Universidad Nacional de Colombia, Bogotá, Colombia
| |
Collapse
|
17
|
Cook PC, Brown SL, Houlder EL, Furlong-Silva J, Conn DP, Colombo SAP, Baker S, Svedberg FR, Howell G, Bertuzzi M, Boon L, Konkel JE, Thornton CR, Allen JE, MacDonald AS. Mgl2 + cDC2s coordinate fungal allergic airway type 2, but not type 17, inflammation in mice. Nat Commun 2025; 16:928. [PMID: 39843887 PMCID: PMC11754877 DOI: 10.1038/s41467-024-55663-3] [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: 01/02/2024] [Accepted: 12/20/2024] [Indexed: 01/24/2025] Open
Abstract
Fungal spores are abundant in the environment and a major cause of asthma. Originally characterised as a type 2 inflammatory disease, allergic airway inflammation that underpins asthma can also involve type 17 inflammation, which can exacerbate disease causing failure of treatments tailored to inhibit type 2 factors. However, the mechanisms that determine the host response to fungi, which can trigger both type 2 and type 17 inflammation in allergic airway disease, remain unclear. Here we find that CD11c+ DCs and CD4+ T cells are essential for development of both type 2 and type 17 airway inflammation in mice repeatedly exposed to inhaled spores. Single cell RNA-sequencing with further multi-parameter cytometry shows that allergic inflammation dramatically alters the proportion of numerous DC clusters in the lung, but that only two of these (Mgl2+ cDC2s and CCR7+ DCs) migrate to the dLNs. Targeted removal of several DC subsets shows that Mgl2+ cDC2 depletion reduces type 2, but not type 17, fungal allergic airway inflammation. These data highlight distinct DC subsets as potential therapeutic targets for the treatment of pulmonary fungal disease.
Collapse
Affiliation(s)
- Peter C Cook
- Medical Research Council Centre for Medical Mycology at the University of Exeter, Department of Biosciences, Faculty of Health and Life Sciences, Geoffrey Pope Building, Stocker Road, Exeter, United Kingdom.
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom.
| | - Sheila L Brown
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Emma L Houlder
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Julio Furlong-Silva
- Medical Research Council Centre for Medical Mycology at the University of Exeter, Department of Biosciences, Faculty of Health and Life Sciences, Geoffrey Pope Building, Stocker Road, Exeter, United Kingdom
| | - Daniel P Conn
- Medical Research Council Centre for Medical Mycology at the University of Exeter, Department of Biosciences, Faculty of Health and Life Sciences, Geoffrey Pope Building, Stocker Road, Exeter, United Kingdom
| | - Stefano A P Colombo
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Syed Baker
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Freya R Svedberg
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Gareth Howell
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Margherita Bertuzzi
- Manchester Fungal Infection Group, University of Manchester, Manchester, United Kingdom
| | | | - Joanne E Konkel
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Christopher R Thornton
- Department of Biosciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, United Kingdom
| | - Judith E Allen
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Andrew S MacDonald
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom.
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom.
| |
Collapse
|
18
|
Kante A, Chevalier MF, Sène D, Chauffier J, Mouly S, Chousterman BG, Azibani F, Terrier B, Pezel T, Comarmond C. Mass cytometry: exploring the immune landscape of systemic autoimmune and inflammatory diseases in the past fourteen years. Front Immunol 2025; 15:1509782. [PMID: 39896815 PMCID: PMC11782038 DOI: 10.3389/fimmu.2024.1509782] [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: 10/11/2024] [Accepted: 12/18/2024] [Indexed: 02/04/2025] Open
Abstract
Auto-immune and inflammatory diseases are heterogenous in their clinical manifestations and prognosis, even among individuals presenting with the same pathology. Understanding the immunological alterations involved in their pathogenesis provides valuable insights in different clinical phenotypes and treatment responses. Immunophenotyping could lead to significant improvements in diagnosis, monitoring, initial treatment decisions and follow-up in autoimmune and inflammatory diseases. Mass cytometry provides measurement of over 40 simultaneous cellular parameters at single-cell resolution, and therefore holds immense potential to evaluate complex cellular systems and for high-dimensional single-cell analysis. The high dimensionality of mass cytometry provides better coverage of immune populations dynamics, with sufficient power to identify rare cell types compared to flow cytometry. In this comprehensive review, we explore how mass cytometry findings contributed in the past decade to a deeper understanding of the cellular actors involved in systemic auto-immune and auto-inflammatory diseases with their respective therapeutic and prognostic impact. We also delve into the bioinformatical approaches applied to mass cytometry to analyze the high volumes of data generated, as well as the impact of the use of complementary single cell RNA sequencing, and their spatial modalities. Our analysis highlights the fact that mass cytometry captures major information on cell populations providing insights on the complex pathogenesis of autoimmune diseases. Future research designs could include mass cytometry findings in association to other -omics to stratify patients in adequate therapeutic arms and provide advancements in personalized therapies in the field of auto-immune and inflammatory diseases.
Collapse
Affiliation(s)
- Aïcha Kante
- Department of Internal Medicine, Lariboisière Hospital, Assistance Publique - Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
- INSERM UMR-S 976, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
| | - Mathieu F. Chevalier
- INSERM UMR-S 976, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
| | - Damien Sène
- Department of Internal Medicine, Lariboisière Hospital, Assistance Publique - Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
- INSERM UMR-S 976, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
| | - Jeanne Chauffier
- Department of Internal Medicine, Lariboisière Hospital, Assistance Publique - Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
- INSERM UMR-S 976, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
| | - Stéphane Mouly
- Department of Internal Medicine, Lariboisière Hospital, Assistance Publique - Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
- INSERM UMR-S 1144, Université Paris Cité, Paris, France
| | - Benjamin Glenn Chousterman
- Department of Anesthesiology and Intensive Care, Lariboisière Hospital, Assistance Publique - Hôpitaux de Paris (AP-HP), Paris, France
- INSERM UMR-S 942 MASCOT - Université Paris Cité, Paris, France
| | - Fériel Azibani
- INSERM UMR-S 942 MASCOT - Université Paris Cité, Paris, France
| | - Benjamin Terrier
- Department of Internal Medicine, Cochin Hospital, Assistance Publique - Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
- INSERM, U970, PARCC, Université de Paris Cité, Paris, France
| | - Théo Pezel
- INSERM UMR-S 942 MASCOT - Université Paris Cité, Paris, France
- Department of Cardiology, Lariboisière Hospital, Université Paris Cité, Paris, France
| | - Cloé Comarmond
- Department of Internal Medicine, Lariboisière Hospital, Assistance Publique - Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
- INSERM UMR-S 976, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
| |
Collapse
|
19
|
Chen Q, Zhao B, Tan Z, Hedberg G, Wang J, Gonzalez L, Mugabo CH, Johnsson A, Negrini E, Páez LP, Rodriguez L, James A, Chen Y, Mikeš J, Bernhardsson AK, Reitzner SM, von Walden F, O'Neill O, Barcenilla H, Wang C, Davis MM, Carlson LM, Pal N, Blomgren K, Repsilber D, Herold N, Lakshmikanth T, Kogner P, Ljungblad L, Brodin P. Systems-level immunomonitoring in children with solid tumors to enable precision medicine. Cell 2025:S0092-8674(24)01427-2. [PMID: 39837329 DOI: 10.1016/j.cell.2024.12.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 10/20/2024] [Accepted: 12/11/2024] [Indexed: 01/23/2025]
Abstract
Cancer is the leading cause of death from disease in children. Survival depends not only on surgery, cytostatic drugs, and radiation but also on systemic immune responses. Factors influencing these immune responses in children of different ages and tumor types are unknown. Novel immunotherapies can enhance anti-tumor immune responses, but few children have benefited, and markers of effective responses are lacking. Here, we present a systems-level analysis of immune responses in 191 children within a population-based cohort with diverse tumors and reveal that age and tumor type shape immune responses differently. Systemic inflammation and cytotoxic T cell responses correlate with tumor mutation rates and immune cell infiltration. Clonally expanded T cell responses are rarely detected in blood or tumors at diagnosis but are sometimes elicited during treatment. Expanded T cells are similarly regulated in children and adults with more immunogenic cancers. This research aims to facilitate the development of precision immunotherapies for children with cancer.
Collapse
Affiliation(s)
- Qi Chen
- Clinical Pediatrics Unit, Department of Women's and Children's Health, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Binbin Zhao
- Clinical Pediatrics Unit, Department of Women's and Children's Health, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Ziyang Tan
- Clinical Pediatrics Unit, Department of Women's and Children's Health, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Gustav Hedberg
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Jun Wang
- Clinical Pediatrics Unit, Department of Women's and Children's Health, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Laura Gonzalez
- Clinical Pediatrics Unit, Department of Women's and Children's Health, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Constantin Habimana Mugabo
- Clinical Pediatrics Unit, Department of Women's and Children's Health, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Anette Johnsson
- Clinical Pediatrics Unit, Department of Women's and Children's Health, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Erika Negrini
- Clinical Pediatrics Unit, Department of Women's and Children's Health, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Laura Piñero Páez
- Clinical Pediatrics Unit, Department of Women's and Children's Health, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Lucie Rodriguez
- Clinical Pediatrics Unit, Department of Women's and Children's Health, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Anna James
- Clinical Pediatrics Unit, Department of Women's and Children's Health, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Yang Chen
- Clinical Pediatrics Unit, Department of Women's and Children's Health, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Jaromír Mikeš
- Clinical Pediatrics Unit, Department of Women's and Children's Health, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Anna Karin Bernhardsson
- Clinical Pediatrics Unit, Department of Women's and Children's Health, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Stefan Markus Reitzner
- Clinical Pediatrics Unit, Department of Women's and Children's Health, Karolinska Institutet, 17165 Stockholm, Sweden; Department of Physiology and Pharmacology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Ferdinand von Walden
- Neuropediatric Unit, Department of Women's and Children's Health, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Olivia O'Neill
- Department of Immunology and Inflammation, Imperial College London, London W12 EH7, UK; Medical Research Council, Laboratory of Medical Sciences, Imperial College Hammersmith Campus, London, UK
| | - Hugo Barcenilla
- Clinical Pediatrics Unit, Department of Women's and Children's Health, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Chunlin Wang
- Institute of Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Mark M Davis
- Institute of Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Lena-Maria Carlson
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, 17177 Stockholm, Sweden; Section Pediatric Oncology, Astrid Lindgren Children's Hospital, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Niklas Pal
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, 17177 Stockholm, Sweden; Section Pediatric Oncology, Astrid Lindgren Children's Hospital, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Klas Blomgren
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, 17177 Stockholm, Sweden; Section Pediatric Oncology, Astrid Lindgren Children's Hospital, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Dirk Repsilber
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Nikolas Herold
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, 17177 Stockholm, Sweden; Section Pediatric Oncology, Astrid Lindgren Children's Hospital, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Tadepally Lakshmikanth
- Clinical Pediatrics Unit, Department of Women's and Children's Health, Karolinska Institutet, 17165 Stockholm, Sweden; Department of Immunology and Inflammation, Imperial College London, London W12 EH7, UK; Medical Research Council, Laboratory of Medical Sciences, Imperial College Hammersmith Campus, London, UK
| | - Per Kogner
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, 17177 Stockholm, Sweden; Section Pediatric Oncology, Astrid Lindgren Children's Hospital, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Linda Ljungblad
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Petter Brodin
- Clinical Pediatrics Unit, Department of Women's and Children's Health, Karolinska Institutet, 17165 Stockholm, Sweden; Department of Immunology and Inflammation, Imperial College London, London W12 EH7, UK; Medical Research Council, Laboratory of Medical Sciences, Imperial College Hammersmith Campus, London, UK; Pediatric Rheumatology, Astrid Lindgren Children's Hospital, Karolinska University Hospital, 17176 Stockholm, Sweden.
| |
Collapse
|
20
|
Yildiz O, Hunt GP, Schroth J, Dhillon G, Spargo TP, Al-Chalabi A, Koks S, Turner MR, Shaw PJ, Henson SM, Iacoangeli A, Malaspina A. Lipid-mediated resolution of inflammation and survival in amyotrophic lateral sclerosis. Brain Commun 2025; 7:fcae402. [PMID: 39816195 PMCID: PMC11733686 DOI: 10.1093/braincomms/fcae402] [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: 04/03/2024] [Revised: 07/26/2024] [Accepted: 01/10/2025] [Indexed: 01/18/2025] Open
Abstract
Neuroinflammation impacts on the progression of amyotrophic lateral sclerosis (ALS), a fatal neurodegenerative disorder. Specialized pro-resolving mediators trigger the resolution of inflammation. We investigate the specialized pro-resolving mediator blood profile and their receptors' expression in peripheral blood mononuclear cells in relation to survival in ALS. People living with ALS (pwALS) were stratified based on bulbar versus limb onset and on key progression metrics using a latent class model, to separate faster progressing from slower progressing ALS. Specialized pro-resolving mediator blood concentrations were measured at baseline and in one additional visit in 20 pwALS and 10 non-neurological controls (Cohort 1). Flow cytometry was used to study the GPR32 and GPR18 resolvin receptors' expression in peripheral blood mononuclear cells from 40 pwALS and 20 non-neurological controls (Cohort 2) at baseline and in two additional visits in 17 pwALS. Survival analysis was performed using Cox proportional hazards models, including known clinical predictors and GPR32 and GPR18 mononuclear cell expression. Differential expression and linear discriminant analyses showed that plasma resolvins were able to distinguish phenotypic variants of ALS from non-neurological controls. RvE3 was elevated in blood from pwALS, whilst RvD1, RvE3, RvT4 and RvD1n-3 DPA were upregulated in A-S and RvD2 in A-F. Compared to non-neurological controls, GPR32 was upregulated in monocytes expressing the active inflammation-suppressing CD11b+ integrin from fast-progressing pwALS, including those with bulbar onset disease (P < 0.0024), whilst GPR32 and GPR18 were downregulated in most B and T cell subtypes. Only GPR18 was upregulated in naïve double positive Tregs, memory cytotoxic Tregs, senescent late memory B cells and late senescent CD8+ T cells from pwALS compared to non-neurological controls (P < 0.0431). Higher GPR32 and GPR18 median expression in blood mononuclear cells was associated with longer survival, with GPR32 expression in classical monocytes (hazard ratio: 0.11, P = 0.003) and unswitched memory B cells (hazard ratio: 0.44, P = 0.008) showing the most significant association, along with known clinical predictors. Low levels of resolvins and downregulation of their membrane receptors in blood mononuclear cells are linked to a faster progression of ALS. Higher mononuclear cell expression of resolvin receptors is a predictor of longer survival. These findings suggest a lipid-mediated neuroprotective response that could be harnessed to develop novel therapeutic strategies and biomarkers for ALS.
Collapse
Affiliation(s)
- Ozlem Yildiz
- Neuromuscular Department, Motor Neuron Disease Centre, Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
- Neuroscience and Trauma, The Blizard Institute, Queen Mary University of London, London E1 2AT, UK
| | - Guy P Hunt
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9RT, UK
- Perron Institute for Neurological and Translational Science, Research Institute in Nedlands, WA 6009, Australia
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth 6150, Australia
| | - Johannes Schroth
- Translational Medicine and Therapeutics, William Harvey Research Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Gurleen Dhillon
- Neuroscience and Trauma, The Blizard Institute, Queen Mary University of London, London E1 2AT, UK
| | - Thomas P Spargo
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9RT, UK
| | - Ammar Al-Chalabi
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9RT, UK
- Maurice Wohl Clinical Neuroscience Institute, King’s College Hospital, London SE5 9RS, UK
| | - Sulev Koks
- Perron Institute for Neurological and Translational Science, Research Institute in Nedlands, WA 6009, Australia
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth 6150, Australia
| | - Martin R Turner
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 7JX, UK
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Sian M Henson
- Translational Medicine and Therapeutics, William Harvey Research Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Alfredo Iacoangeli
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9RT, UK
- National Institute for Health Research Biomedical Research Centre and Dementia Unit at South London and Maudsley NHS Foundation Trust and King’s College London, London SE5 8AF, UK
| | - Andrea Malaspina
- Neuromuscular Department, Motor Neuron Disease Centre, Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
- Neuroscience and Trauma, The Blizard Institute, Queen Mary University of London, London E1 2AT, UK
| |
Collapse
|
21
|
Caldwell BA, Ie S, Lucas A, Li L. Ticam2 ablation facilitates monocyte exhaustion recovery after sepsis. Sci Rep 2025; 15:2059. [PMID: 39814939 PMCID: PMC11735619 DOI: 10.1038/s41598-025-86103-x] [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: 09/10/2024] [Accepted: 01/08/2025] [Indexed: 01/18/2025] Open
Abstract
Sepsis is a leading cause of death worldwide, with most patient mortality stemming from lingering immunosuppression in sepsis survivors. This is due in part to immune dysfunction resulting from monocyte exhaustion, a phenotype of reduced antigen presentation, altered CD14/CD16 inflammatory subtypes, and disrupted cytokine production. Whereas previous research demonstrated improved sepsis survival in Ticam2-/- mice, the contribution of TICAM2 to long-term exhaustion memory remained unknown. Using a cecal slurry injection sepsis model, we monitored the establishment and recovery of monocyte exhaustion in Ticam2-/- mice. After one week of recovery, we profiled bone marrow and splenic reservoir monocytes in Ticam2-/- mice and found that, in contrast to the persistent exhaustion observed in wild-type monocytes, Ticam2-/- monocytes largely resembled healthy controls. To determine the impact of TICAM2 ablation on innate epigenetic memory in sepsis, we measured genome-wide DNA methylation in bone marrow monocytes and found that Ticam2-/- cells exhibit a unique profile of altered methylation at CEBPE binding sites and regulatory features for key immune genes such as Dmkn and Btg1. Bearing human translational relevance, a case study of time course blood samples collected from a sepsis patient presenting with SIRS and a positive qSOFA revealed a similar effect in human monocytes, which steadily transition into an exhausted memory characterized by a CD38high; CX3CR1low; HLA-DRlow state within four days of hospital admittance. Together, our data reveal the chronic preservation of monocyte exhaustion, partially controlled by TICAM2.
Collapse
Affiliation(s)
- Blake A Caldwell
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, 24061-0910, USA
| | - Susanti Ie
- Carillion Roanoke Memorial Hospital, Roanoke, VA, 24014, USA
| | - Amy Lucas
- Carillion Roanoke Memorial Hospital, Roanoke, VA, 24014, USA
| | - Liwu Li
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, 24061-0910, USA.
| |
Collapse
|
22
|
Shirasawa M, Yoshida T, Matsutani T, Takeyasu Y, Goto N, Yagishita S, Kitano S, Kuroda H, Hida T, Kurata T, Ohe Y. Diversity of TCR repertoire predicts recurrence after CRT followed by durvalumab in patients with NSCLC. NPJ Precis Oncol 2025; 9:17. [PMID: 39814862 PMCID: PMC11735630 DOI: 10.1038/s41698-024-00781-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 12/10/2024] [Indexed: 01/18/2025] Open
Abstract
Chemoradiotherapy (CRT) followed by durvalumab is standard for unresectable locally advanced non-small-cell lung cancer (LA-NSCLC). This study assesses how CRT alters the T-cell receptor (TCR) repertoire in CD8 + PD-1 + T-cells and its impact on clinical outcomes. This prospective study, conducted from November 2019 to May 2021 at three institutions in Japan, evaluated the diversity of TCR repertoire (DE50) in PD-1 + CD8 + T-cells and CD8 + T-cell phenotypes in peripheral blood before and after CRT. Forty patients treated with CRT were included. The diversity and usage of TCR beta variable chains (TRBV) and 14 junctional chains (TRBJ) were significantly and positively correlated before and after CRT. Regarding the DE50, the progression-free survival (PFS) of patients with DE50High before CRT was significantly greater than that of those with DE50Low (NR vs. NR months, HR 0.17, p = 0.01). The diversity of TCR repertoire might more accurately predict the efficacy of CRT followed by durvalumab therapy.
Collapse
Affiliation(s)
- Masayuki Shirasawa
- Department of Thoracic Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
- Department of Respiratory Medicine, Kitasato University School of Medicine, 1-15-1, Kitasato, Minami-ku, Sagamihara city, Kanagawa, 252-0375, Japan
| | - Tatsuya Yoshida
- Department of Thoracic Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.
- Department of Experimental Therapeutics, National Cancer Center Research Institute, 5-1-1, Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.
| | - Takaji Matsutani
- Repertoire Genesis, Inc, Ibaraki, Osaka, Japan
- Translational Research Department, Maruho Co. Ltd., Kyoto, Japan
| | - Yuki Takeyasu
- Department of Thoracic Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
- Department of Thoracic Oncology, Kansai Medical University Hospital, 2-3-1 Shinmachi, Hirakata, Osaka, 573-1191, Japan
| | - Naoko Goto
- Department of Thoracic Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Shigehiro Yagishita
- Division of Molecular Pharmacology, National Cancer Center Research Institute, 5-1-1, Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Shigehisa Kitano
- Division of Cancer Immunotherapy Development, Department of Advanced Medical Development, The Cancer Institute Hospital of JFCR, 3-8-31, Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Hiroaki Kuroda
- Department of Thoracic Surgery, Aichi Cancer Center Hospital, 1-1 Kanokoden, Chikusa-ku, Nagoya, Aichi, 464-8681, Japan
| | - Toyoaki Hida
- Department of Thoracic Oncology, Aichi Cancer Center Hospital, 1-1 Kanokoden, Chikusa-ku, Nagoya, Aichi, 464-8681, Japan
| | - Takayasu Kurata
- Department of Thoracic Oncology, Kansai Medical University Hospital, 2-3-1 Shinmachi, Hirakata, Osaka, 573-1191, Japan
| | - Yuichiro Ohe
- Department of Thoracic Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| |
Collapse
|
23
|
Saleem SK, Decker S, Kissel S, Bauer M, Chernyakov D, Bräuer-Hartmann D, Aumann K, Wickenhauser C, Herling M, Skorobohatko O, Mathew N, Schmidt C, Klein C, Follo M, Dierks C. JNK1 inhibitors target distal B cell receptor signaling and overcome BTK-inhibitor resistance in CLL. J Exp Med 2025; 222:e20230681. [PMID: 39570282 PMCID: PMC11586660 DOI: 10.1084/jem.20230681] [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: 04/21/2023] [Revised: 03/21/2024] [Accepted: 07/08/2024] [Indexed: 11/22/2024] Open
Abstract
Inhibition of the proximal B cell receptor (BCR) signaling pathway by BTK inhibitors is highly effective in the treatment of CLL, but drug resistance or intolerance occurs. Here, we investigated c-Jun N-terminal protein kinase 1 (JNK1) as an alternative drug target in the distal BCR pathway. JNK1 was preferentially overexpressed and activated in poor prognostic CLL with unmutated IGHV. Proximal BCR inhibition (BTK, PI3K, or SYK inhibitors) or SYK knockdown efficiently dephosphorylated JNK1, identifying JNK1 as a critical BCR downstream kinase in CLL. JNK1 inhibition induced apoptosis in primary CLL cells, resulting in the downregulation of BCL2, MCL1, and c-JUN. JNK1 inhibition in patient-derived CLL xenografted mice and Eµ-TCL1-tg mice prevented CLL progression, reduced splenic infiltration, and restored T cell function and normal hematopoiesis. JNK1 inhibitors even remained effective in ibrutinib refractory CLL. In conclusion, our study revealed JNK1 as a promising drug target in CLL downstream of the BCR, overcoming ibrutinib resistance, blocking the protective microenvironment, and improving CLL-specific immunosuppressive mechanisms.
Collapse
MESH Headings
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Humans
- Animals
- Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors
- Agammaglobulinaemia Tyrosine Kinase/metabolism
- Receptors, Antigen, B-Cell/metabolism
- Drug Resistance, Neoplasm/drug effects
- Signal Transduction/drug effects
- Mice
- Mitogen-Activated Protein Kinase 8/metabolism
- Mitogen-Activated Protein Kinase 8/genetics
- Mitogen-Activated Protein Kinase 8/antagonists & inhibitors
- Adenine/analogs & derivatives
- Adenine/pharmacology
- Protein Kinase Inhibitors/pharmacology
- Piperidines/pharmacology
- Apoptosis/drug effects
- Female
- Pyrimidines/pharmacology
- Syk Kinase/metabolism
- Syk Kinase/antagonists & inhibitors
- Male
- Mice, Transgenic
Collapse
Affiliation(s)
- Shifa Khaja Saleem
- Department of Hematology and Oncology, Freiburg University Medical Center, Albert-Ludwigs-University of Freiburg, Breisgau, Germany
| | - Sarah Decker
- Department of Hematology and Oncology, Freiburg University Medical Center, Albert-Ludwigs-University of Freiburg, Breisgau, Germany
| | - Sandra Kissel
- Department of Hematology and Oncology, Freiburg University Medical Center, Albert-Ludwigs-University of Freiburg, Breisgau, Germany
| | - Marcus Bauer
- Institute of Pathology, University Hospital Halle, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Dmitry Chernyakov
- Department of Hematology/Oncology and Stem Cell Transplantation, KIM IV, Martin-Luther University Halle-Wittenberg, Halle, Germany
| | - Daniela Bräuer-Hartmann
- Department of Hematology/Oncology and Stem Cell Transplantation, KIM IV, Martin-Luther University Halle-Wittenberg, Halle, Germany
| | - Konrad Aumann
- Department of Pathology, Freiburg University Medical Center, Albert-Ludwigs-University of Freiburg, Breisgau, Germany
| | - Claudia Wickenhauser
- Institute of Pathology, University Hospital Halle, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Marco Herling
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectious Diseases, University of Leipzig, Leipzig, Germany
| | - Oleksandra Skorobohatko
- Department of Hematology/Oncology and Stem Cell Transplantation, KIM IV, Martin-Luther University Halle-Wittenberg, Halle, Germany
| | - Nimitha Mathew
- Department of Hematology and Oncology, Freiburg University Medical Center, Albert-Ludwigs-University of Freiburg, Breisgau, Germany
| | - Cornelius Schmidt
- Department of Hematology and Oncology, Freiburg University Medical Center, Albert-Ludwigs-University of Freiburg, Breisgau, Germany
| | - Claudius Klein
- Department of Hematology and Oncology, Freiburg University Medical Center, Albert-Ludwigs-University of Freiburg, Breisgau, Germany
- Department of Nuclear Medicine, Freiburg University Medical Center, Albert-Ludwigs-University of Freiburg, Breisgau, Germany
| | - Marie Follo
- Department of Hematology and Oncology, Freiburg University Medical Center, Albert-Ludwigs-University of Freiburg, Breisgau, Germany
| | - Christine Dierks
- Department of Hematology and Oncology, Freiburg University Medical Center, Albert-Ludwigs-University of Freiburg, Breisgau, Germany
- Department of Hematology/Oncology and Stem Cell Transplantation, KIM IV, Martin-Luther University Halle-Wittenberg, Halle, Germany
| |
Collapse
|
24
|
Lee Y, Chen ELY, Chan DCH, Dinesh A, Afiuni-Zadeh S, Klamann C, Selega A, Mrkonjic M, Jackson HW, Campbell KR. Segmentation aware probabilistic phenotyping of single-cell spatial protein expression data. Nat Commun 2025; 16:389. [PMID: 39755686 DOI: 10.1038/s41467-024-55214-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 12/02/2024] [Indexed: 01/06/2025] Open
Abstract
Spatial protein expression technologies can map cellular content and organization by simultaneously quantifying the expression of >40 proteins at subcellular resolution within intact tissue sections and cell lines. However, necessary image segmentation to single cells is challenging and error prone, easily confounding the interpretation of cellular phenotypes and cell clusters. To address these limitations, we present STARLING, a probabilistic machine learning model designed to quantify cell populations from spatial protein expression data while accounting for segmentation errors. To evaluate performance, we develop a comprehensive benchmarking workflow by generating highly multiplexed imaging data of cell line pellet standards with controlled cell content and marker expression and additionally established a score to quantify the biological plausibility of discovered cellular phenotypes on patient-derived tissue sections. Moreover, we generate spatial expression data of the human tonsil-a densely packed tissue prone to segmentation errors-and demonstrate cellular states captured by STARLING identify known cell types not visible with other methods and enable quantification of intra- and inter- individual heterogeneity.
Collapse
Affiliation(s)
- Yuju Lee
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
- Department of Computer Science, University of Toronto, Toronto, ON, Canada
| | - Edward L Y Chen
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Darren C H Chan
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Anuroopa Dinesh
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Somaieh Afiuni-Zadeh
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Conor Klamann
- Data Sciences Institute, University of Toronto, Toronto, ON, Canada
| | - Alina Selega
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
- Vector Institute, Toronto, ON, Canada
| | - Miralem Mrkonjic
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Hartland W Jackson
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
- Ontario Institute of Cancer Research, Toronto, ON, Canada.
| | - Kieran R Campbell
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada.
- Department of Computer Science, University of Toronto, Toronto, ON, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
- Vector Institute, Toronto, ON, Canada.
- Ontario Institute of Cancer Research, Toronto, ON, Canada.
- Department of Statistical Sciences, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
25
|
Peters JM, Irvine EB, Makatsa MS, Rosenberg JM, Wadsworth MH, Hughes TK, Sutton MS, Nyquist SK, Bromley JD, Mondal R, Roederer M, Seder RA, Darrah PA, Alter G, Seshadri C, Flynn JL, Shalek AK, Fortune SM, Bryson BD. High-dose intravenous BCG vaccination induces enhanced immune signaling in the airways. SCIENCE ADVANCES 2025; 11:eadq8229. [PMID: 39742484 PMCID: PMC11694782 DOI: 10.1126/sciadv.adq8229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 11/20/2024] [Indexed: 01/03/2025]
Abstract
Intradermal Bacillus Calmette-Guérin (BCG) is the most widely administered vaccine, but it does not sufficiently protect adults against pulmonary tuberculosis. Recent studies in nonhuman primates show that intravenous BCG administration offers superior protection against Mycobacterium tuberculosis (Mtb). We used single-cell analysis of bronchoalveolar lavage cells from rhesus macaques vaccinated via different routes and doses of BCG to identify alterations in the immune ecosystem in the airway following vaccination. Our findings reveal that high-dose intravenous BCG induces an influx of polyfunctional T cells and macrophages in the airways, with alveolar macrophages from high-dose intravenous BCG displaying a basal activation state in the absence of purified protein derivative stimulation, defined in part by interferon signaling. Enhanced intercellular immune signaling and stronger T helper 1-T helper 17 transcriptional responses were observed following purified protein derivative stimulation. These results suggest that high-dose intravenous BCG vaccination creates a specialized immune environment that primes airway cells for effective Mtb clearance.
Collapse
Affiliation(s)
- Joshua M. Peters
- Department of Biological Engineering, MIT, Cambridge, MA, USA
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Edward B. Irvine
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Mohau S. Makatsa
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Jacob M. Rosenberg
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Division of Infectious Diseases, MGH, Boston, MA, USA
| | - Marc H. Wadsworth
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering & Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
| | - Travis K. Hughes
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering & Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
| | | | - Sarah K. Nyquist
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering & Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
| | - Joshua D. Bromley
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering & Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
| | - Rajib Mondal
- Research Laboratory of Electronics, Harvard-MIT Program in Health Sciences and Technology, Cambridge, MA, USA
| | | | | | | | - Galit Alter
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA, USA
| | - Chetan Seshadri
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - JoAnne L. Flynn
- Department of Microbiology and Molecular Genetics and Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Alex K. Shalek
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering & Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
| | - Sarah M. Fortune
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Bryan D. Bryson
- Department of Biological Engineering, MIT, Cambridge, MA, USA
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA, USA
| |
Collapse
|
26
|
Kuett L, Bollhagen A, Tietscher S, Sobottka B, Eling N, Varga Z, Moch H, de Souza N, Bodenmiller B. Distant Metastases of Breast Cancer Resemble Primary Tumors in Cancer Cell Composition but Differ in Immune Cell Phenotypes. Cancer Res 2025; 85:15-31. [PMID: 39437149 DOI: 10.1158/0008-5472.can-24-1211] [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: 04/17/2024] [Revised: 07/02/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024]
Abstract
Breast cancer is the most commonly diagnosed cancer in women, with distant metastasis being the main cause of breast cancer-related deaths. Elucidating the changes in the tumor and immune ecosystems that are associated with metastatic disease is essential to improve understanding and ultimately treatment of metastasis. Here, we developed an in-depth, spatially resolved single-cell atlas of the phenotypic diversity of tumor and immune cells in primary human breast tumors and matched distant metastases, using imaging mass cytometry to analyze a total of 75 unique antibody targets. Although the same tumor cell phenotypes were typically present in primary tumors and metastatic sites, suggesting a strong founder effect of the primary tumor, their proportions varied between matched samples. Notably, the metastatic site did not influence tumor phenotype composition, except for the brain. Metastatic sites exhibited a lower number of immune cells overall but had a higher proportion of myeloid cells as well as exhausted and cytotoxic T cells. Myeloid cells showed distinct tissue-specific compositional signatures and increased presence of potentially matrix remodeling phenotypes in metastatic sites. This analysis of tumor and immune cell phenotypic composition of metastatic breast cancer highlights the heterogeneity of the disease within patients and across distant metastatic sites, indicating myeloid cells as the predominant immune modulators that could potentially be targeted at these sites. Significance: Multiplex imaging analysis of matched primary and metastatic breast tumors provides a phenotypic and spatial map of tumor microenvironments, revealing similar compositions of cancer cells and divergent immunologic features between matched samples.
Collapse
Affiliation(s)
- Laura Kuett
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
- Life Science Zurich Graduate School, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Alina Bollhagen
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
- Life Science Zurich Graduate School, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Sandra Tietscher
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
- Life Science Zurich Graduate School, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Bettina Sobottka
- Department of Pathology and Molecular Pathology, University and University Hospital Zurich, Zurich, Switzerland
| | - Nils Eling
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Zsuzsanna Varga
- Department of Pathology and Molecular Pathology, University and University Hospital Zurich, Zurich, Switzerland
| | - Holger Moch
- Department of Pathology and Molecular Pathology, University and University Hospital Zurich, Zurich, Switzerland
| | - Natalie de Souza
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Bernd Bodenmiller
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
27
|
Zhan D, Du Z, Zhang S, Huang J, Zhang J, Zhang H, Liu Z, Menu E, Wang J. Targeting Caveolin-1 in Multiple Myeloma Cells Enhances Chemotherapy and Natural Killer Cell-Mediated Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408373. [PMID: 39630017 PMCID: PMC11789597 DOI: 10.1002/advs.202408373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 11/18/2024] [Indexed: 01/30/2025]
Abstract
The cell membrane transport capacity and surface targets of multiple myeloma (MM) cells heavily influence chemotherapy and immunotherapy. Here, it is found that caveolin-1 (CAV1), a primary component of membrane lipid rafts and caveolae, is highly expressed in MM cells and is associated with MM progression and drug resistance. CAV1 knockdown decreases MM cell adhesion to stromal cells and attenuates cell adhesion-mediated drug resistance to bortezomib. CAV1 inhibition in MM cells enhances natural killer cell-mediated cytotoxicity through increasing CXCL10, SLAMF7, and CD112. CAV1 suppression reduces mitochondrial membrane potential, increases reactive oxygen species, and inhibits autophagosome-lysosome fusion, resulting in the disruption of redox homeostasis. Additionally, CAV1 knockdown enhances glutamine addiction by increasing ASCT2 and LAT1 and dysregulates glutathione metabolism. As a result of CAV1 inhibition, MM cells are more sensitive to starvation, glutamine depletion, and glutamine transporter inhibition, and grow more slowly in vivo in a mouse model treated with bortezomib. The observation that CAV1 inhibition modulated by 6-mercaptopurine, daidzin, and statins enhances the efficacy of bortezomib in vitro and in vivo highlights the translational significance of these FDA-approved drugs in improving MM outcomes. These data demonstrate that CAV1 serves as a potent therapeutic target for enhancing chemotherapy and immunotherapy for MM.
Collapse
Affiliation(s)
- Dewen Zhan
- The Affiliated Traditional Chinese Medicine HospitalGuangzhou Medical UniversityGuangzhou510130China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and DegradationSchool of Basic Medical SciencesGuangzhou Medical UniversityGuangzhou511436China
| | - Zhimin Du
- School of NursingGuangzhou Medical UniversityGuangzhou510182China
| | - Shang Zhang
- The Affiliated Traditional Chinese Medicine HospitalGuangzhou Medical UniversityGuangzhou510130China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and DegradationSchool of Basic Medical SciencesGuangzhou Medical UniversityGuangzhou511436China
| | - Juanru Huang
- The Affiliated Traditional Chinese Medicine HospitalGuangzhou Medical UniversityGuangzhou510130China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and DegradationSchool of Basic Medical SciencesGuangzhou Medical UniversityGuangzhou511436China
| | - Jian Zhang
- School of Biomedical EngineeringGuangzhou Medical UniversityGuangzhou511436China
| | - Hui Zhang
- The Affiliated Traditional Chinese Medicine HospitalGuangzhou Medical UniversityGuangzhou510130China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and DegradationSchool of Basic Medical SciencesGuangzhou Medical UniversityGuangzhou511436China
| | - Zhongrui Liu
- The Affiliated Traditional Chinese Medicine HospitalGuangzhou Medical UniversityGuangzhou510130China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and DegradationSchool of Basic Medical SciencesGuangzhou Medical UniversityGuangzhou511436China
| | - Eline Menu
- Department of Hematology and ImmunologyMyeloma Center BrusselsVrije Universiteit BrusselBrusselsB‐1090Belgium
| | - Jinheng Wang
- The Affiliated Traditional Chinese Medicine HospitalGuangzhou Medical UniversityGuangzhou510130China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and DegradationSchool of Basic Medical SciencesGuangzhou Medical UniversityGuangzhou511436China
| |
Collapse
|
28
|
Dowrey TW, Cranston SF, Skvir N, Lok Y, Gould B, Petrowitz B, Villar D, Shan J, James M, Dodge M, Belkina AC, Giadone RM, Milman S, Sebastiani P, Perls TT, Andersen SL, Murphy GJ. A longevity-specific bank of induced pluripotent stem cells from centenarians and their offspring. Aging Cell 2025; 24:e14351. [PMID: 39319670 PMCID: PMC11709102 DOI: 10.1111/acel.14351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 08/02/2024] [Accepted: 09/05/2024] [Indexed: 09/26/2024] Open
Abstract
Centenarians provide a unique lens through which to study longevity, healthy aging, and resiliency. Moreover, models of human aging and resilience to disease that allow for the testing of potential interventions are virtually non-existent. We obtained and characterized over 96 centenarian and offspring peripheral blood samples including those connected to functional independence data highlighting resistance to disability and cognitive impairment. Targeted methylation arrays were used in molecular aging clocks to compare and contrast differences between biological and chronological age in these specialized subjects. Isolated peripheral blood mononuclear cells (PBMCs) from 20 of these subjects were then successfully reprogrammed into high-quality induced pluripotent stem cell (iPSC) lines which were functionally characterized for pluripotency, genomic stability, and the ability to undergo directed differentiation. The result of this work is a one-of-a-kind resource for studies of human longevity and resilience that can fuel the discovery and validation of novel therapeutics for aging-related disease.
Collapse
Affiliation(s)
- Todd W. Dowrey
- Center for Regenerative Medicine of Boston University and Boston Medical CenterBostonMassachusettsUSA
- Section of Hematology and Medical OncologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | - Samuel F. Cranston
- Center for Regenerative Medicine of Boston University and Boston Medical CenterBostonMassachusettsUSA
- Section of Hematology and Medical OncologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | - Nicholas Skvir
- Center for Regenerative Medicine of Boston University and Boston Medical CenterBostonMassachusettsUSA
- Section of Hematology and Medical OncologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | - Yvonne Lok
- Center for Regenerative Medicine of Boston University and Boston Medical CenterBostonMassachusettsUSA
- Section of Hematology and Medical OncologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | - Brian Gould
- Section of Geriatrics, Department of MedicineBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | - Bradley Petrowitz
- Section of Geriatrics, Department of MedicineBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | | | - Jidong Shan
- Albert Einstein College of MedicineBronxNew YorkUSA
| | - Marianne James
- Center for Regenerative Medicine of Boston University and Boston Medical CenterBostonMassachusettsUSA
| | - Mark Dodge
- Center for Regenerative Medicine of Boston University and Boston Medical CenterBostonMassachusettsUSA
| | - Anna C. Belkina
- Flow Cytometry Core FacilityBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Department of Pathology and Laboratory MedicineBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | - Richard M. Giadone
- Department of Stem Cell and Regenerative BiologyHarvard UniversityCambridgeMassachusettsUSA
| | | | - Paola Sebastiani
- Institute for Clinical Research and Health Policy StudiesTufts Medical CenterBostonMassachusettsUSA
| | - Thomas T. Perls
- Section of Geriatrics, Department of MedicineBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | - Stacy L. Andersen
- Section of Geriatrics, Department of MedicineBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | - George J. Murphy
- Center for Regenerative Medicine of Boston University and Boston Medical CenterBostonMassachusettsUSA
- Section of Hematology and Medical OncologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| |
Collapse
|
29
|
Biwott K, Singh P, Baráth S, Nyariki JN, Hevessy Z, Bacso Z. Dynamic P-glycoprotein expression in early and late memory states of human CD8 + T cells and the protective role of ruxolitinib. Biomed Pharmacother 2025; 182:117780. [PMID: 39740391 DOI: 10.1016/j.biopha.2024.117780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/14/2024] [Accepted: 12/20/2024] [Indexed: 01/02/2025] Open
Abstract
ABCB1/MDR-1/P-glycoprotein (Pgp) is an ABC transporter responsible for cancer cell multi-drug resistance. It is expressed in cytotoxic T lymphocytes (CTL). Eliminating sensitive cancer cells during high-dose chemotherapy can also damage immune cells. Our study aimed to assess which maturing human CD8 + CTL memory subsets may be affected based on their Pgp protein expression. In an in vitro CTL differentiation model system, we tracked the maturation of naive, effector, and memory cells and the expression of Pgp. This system involves co-culturing blood lymphocytes with proliferation-inhibited JY antigen-presenting B-lymphoblastoid cells expressing HLA-I A2. These JY-primed maturing CTLs were TCR-activated using beads, and the effect of the maturation-modifying JAK1/2 inhibitor ruxolitinib was examined. Multidimensional analysis identified six major CTL subsets: naive, young memory (Tym), stem cell memory (Tscm), central memory (Tcm), effector memory (Tem), and effectors (Te). These subsets were further divided into thirteen specific subsets: TymCD127 + , TymCD127-, Tscm, TcmCD95 + , TcmCD73 +CD95 + , TcmCD95+CD127 + , TcmPD1 + , TemCD95 + , TemraCD127 + , TemraCD127-, TeCD95 + , and TeCD73 +CD95 + . Pgp expression was detectable in naïve cells and dynamically changed across the thirteen identified subsets. Increased Pgp was detected in young memory T cells and in Tscm, TcmCD95 + , and TcmPD1 + human CTL subsets. Unlike other transiently appearing memory cells, the number of cells in these core Pgp-expressing memory subsets stabilized by the end of the contraction phase. Ruxolitinib treatment downregulated effector T-cell polarization while upregulating small memory subsets expressing Pgp. In conclusion, activation increased Pgp expression, whereas ruxolitinib treatment preserved small early and late memory subset core that primarily expressed Pgp.
Collapse
Affiliation(s)
- Kipchumba Biwott
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen 4032, Hungary; Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, Debrecen 4032, Hungary; Department of Biochemistry and Biotechnology, Technical University of Kenya, Kenya.
| | - Parvind Singh
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen 4032, Hungary.
| | - Sándor Baráth
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen 4032, Hungary.
| | | | - Zsuzsanna Hevessy
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen 4032, Hungary.
| | - Zsolt Bacso
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen 4032, Hungary; Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, Debrecen 4032, Hungary; Dean's office, Faculty of Pharmacy, University of Debrecen, Debrecen 4032, Hungary.
| |
Collapse
|
30
|
DeCostanza L, Grogan GM, Bruce AC, Peachey CM, Clark EA, Atkins K, Tylek T, Solga MD, Spiller KL, Peirce SM, Campbell CA, Cottler PS. Decellularized porcine dermal hydrogel enhances implant-based wound healing in the setting of irradiation. Acta Biomater 2025; 191:260-275. [PMID: 39522628 DOI: 10.1016/j.actbio.2024.11.009] [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: 08/13/2024] [Revised: 11/04/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
Acellular Dermal Matrix (ADM) provides mechanical and soft tissue support in implant-based breast reconstruction, and has shown to modulate the healing response. However, skin flap necrosis, edema, and previous radiation therapy can hinder ADM integration. Effective biomaterial integration requires regulating the immune response, fibrosis, and adipocyte-driven functionalization. Extracellular matrix (ECM) hydrogels have demonstrated utility in tissue regeneration, and decreasing inflammation and fibrosis in various tissues. Therefore, we hypothesized that a Decellularized Porcine Dermal (DPD) hydrogel to support ADM integration would prevent excessive fibrosis, regulate the macrophage response, and promote adipogenesis. Exploration of DPD hydrogel during ADM implantation in mice (healthy and radiated) revealed long-term effects of irradiation on implant wound healing. DPD hydrogel rescued radiation-induced fibrosis, restoring capsule thickness of healthy mice, and did not increase the fibroblast migration into the ADM. As a modulating soft tissue filler, DPD hydrogel also promoted adipocyte infiltration in healthy and irradiated mice. Detailed macrophage analysis showed that radiation led to the increase in pro-inflammatory, transition, and reparative markers. Despite relatively subtle effects on individual macrophage phenotype markers, multidimensional flow cytometry analysis revealed that DPD hydrogel temporally regulated two subpopulations. he presence of DPD resulted in significantly reduced CD9HiArg1HiCD301bLo and CD163HiCD38HiCD301bHi macrophages in healthy mice at one week, and a significant increase in CD9High macrophages with low expression of other markers at 6 weeks in irradiated mice. DPD hydrogel promotes a decreased fibrotic, and adipocyte-promoting coordination of wound healing in healthy and irradiated wound beds while not disrupting the immunomodulatory effects of ADM. STATEMENT OF SIGNIFICANCE: Acellular Dermal Matrix (ADM) provides mechanical and soft tissue support in post-mastectomy implant-based breast reconstruction, and positively affects wound healing. Following breast reconstruction, skin flap necrosis, edema, and previous radiation therapy can hinder ADM integration. Effective wound healing and biomaterial integration requires regulating the cellular immune response. Extracellular matrix hydrogels have demonstrated utility in tissue regeneration and decreasing inflammation and fibrosis in various tissues, but has yet to be utilized in the setting of breast reconstruction. Here, we demonstrated that a decellularized dermal hydrogel as an adjunct to ADM, decreases fibrosis and promotes adipogenesis during the coordination of wound healing in healthy and clinically relevant microenvironments that have received radiation therapy while not disrupting the immunomodulatory effects of implanted ADM.
Collapse
Affiliation(s)
- Lillian DeCostanza
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Graham M Grogan
- Department of Plastic Surgery, University of Virginia, Charlottesville, VA, USA
| | - Anthony C Bruce
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Corrina M Peachey
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Evan A Clark
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Kristen Atkins
- Department of Pathology, University of Virginia, Charlottesville, VA, USA
| | - Tina Tylek
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Michael D Solga
- Flow Cytometry Core Facility, University of Virginia, Charlottesville, VA, USA
| | - Kara L Spiller
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Shayn M Peirce
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | | | - Patrick S Cottler
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA; Department of Plastic Surgery, University of Virginia, Charlottesville, VA, USA; Department of Otolaryngology, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
31
|
Gonzalez GC, Li CM, Pasolini I, Pete SI, Verheyen C, Vignolo SM, De Toni T, Stock AA, Tomei AA. High-Yield Generation of Glucose-Responsive Pseudoislets From Murine Insulinoma Cells for In Vitro Studies and Longitudinal Monitoring of Graft Survival In Vivo. Cell Transplant 2025; 34:9636897251315123. [PMID: 39881520 PMCID: PMC11780636 DOI: 10.1177/09636897251315123] [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: 11/01/2024] [Revised: 12/14/2024] [Accepted: 01/06/2025] [Indexed: 01/31/2025] Open
Abstract
Compared to primary pancreatic islets, insulinoma cell-derived 3D pseudoislets offer a more accessible, consistent, renewable, and widely applicable model system for optimization and mechanistic studies in type 1 diabetes (T1D). Here, we report a simple and efficient method for generating 3D pseudoislets from MIN6 and NIT-1 murine insulinoma cells. These pseudoislets are homogeneous in size and morphology (~150 µm), exhibit functional glucose-stimulated insulin secretion (GSIS) up to 18 days (NIT-1) enabling long-term studies, are produced in high yield [>35,000 Islet Equivalence from 30 ml culture], and are suitable for both in vitro and in vivo studies, including for encapsulation studies. To enable non-invasive longitudinal monitoring of graft survival in vivo, we transduced NIT-1 cells with green fluorescent protein-luciferase and confirmed comparable morphology, viability, and GSIS to untransduced cells in vitro. After subcutaneous implantation, we show capability to monitor graft survival in immunodeficient mice, recurrence of autoimmunity in non-obese diabetic mice, and allorejection in C57BL/6 mice. Overall, this platform provides an accessible protocol for generating high yields of 3D pseudoislets and non-invasive longitudinal monitoring of graft survival in different models offer advantages over primary islets for optimization and mechanistic studies of β cell biology, drug discovery, T1D pathogenesis and prevention, and β cell transplantation.
Collapse
Affiliation(s)
- Grisell C. Gonzalez
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Chris M. Li
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Ilaria Pasolini
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Sophia I. Pete
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Connor Verheyen
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Sofia M. Vignolo
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Teresa De Toni
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Aaron A. Stock
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Alice A. Tomei
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
32
|
Tessandier N, Elie B, Boué V, Selinger C, Rahmoun M, Bernat C, Grasset S, Groc S, Bedin AS, Beneteau T, Bonneau M, Graf C, Jacobs N, Kamiya T, Kerioui M, Lajoie J, Melki I, Prétet JL, Reyné B, Schlecht-Louf G, Sofonea MT, Supplisson O, Wymant C, Foulongne V, Guedj J, Hirtz C, Picot MC, Reynes J, Tribout V, Tuaillon É, Waterboer T, Segondy M, Bravo IG, Boulle N, Murall CL, Alizon S. Viral and immune dynamics of genital human papillomavirus infections in young women with high temporal resolution. PLoS Biol 2025; 23:e3002949. [PMID: 39836629 PMCID: PMC11750104 DOI: 10.1371/journal.pbio.3002949] [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: 05/29/2024] [Accepted: 11/22/2024] [Indexed: 01/23/2025] Open
Abstract
Human papillomavirus (HPV) infections drive one in 20 new cancer cases, exerting a particularly high burden on women. Most anogenital HPV infections are cleared in less than two years, but the underlying mechanisms that favour persistence in around 10% of women remain largely unknown. Notwithstanding, it is precisely this information that is crucial for improving treatment, screening, and vaccination strategies. To understand viral and immune dynamics in non-persisting HPV infections, we set up an observational longitudinal cohort study with frequent on-site visits for biological sample collection. We enrolled 189 women aged from 18 to 25 and living in the area of Montpellier (France) between 2016 and 2020. We performed 974 on-site visits for a total of 1,619 months of follow-up. We collected data on virus load, local immune cell populations, local concentrations of cytokines, and circulating antibody titres. Using hierarchical Bayesian statistical modelling to simultaneously analyse the data from 164 HPV infections from 76 participants, we show that in two months after infection, HPV viral load in non-persisting infections reaches a plateau that lasts on average for 13 to 20 months (95% credibility interval) and is then followed by a rapid clearance phase. This first description of the dynamics of HPV infections comes with the identification of immune correlates associated with infection clearance, especially gamma-delta T cells and CXCL10 concentration. A limitation of this study on HPV kinetics is that many infection follow-ups are censored. Furthermore, some immune cell populations are difficult to label because cervical immunity is less well characterised than systemic immunity. These results open new perspectives for understanding the frontier between acute and chronic infections, and for controlling HPV-associated diseases, as well as for research on human cancers of infectious origin. Trial Registration: This trial was registered is registered at ClinicalTrials.gov under the ID NCT02946346. This study has been approved by the Comité de Protection des Personnes (CPP) Sud Méditerranée I (reference number 2016-A00712-49); by the Comité Consultatif sur le Traitement de l'Information en matière de Recherche dans le domaine de la Santé (reference number 16.504); by the Commission Nationale Informatique et Libertés (reference number MMS/ABD/ AR1612278, decision number DR-2016-488), by the Agence Nationale de Sécurité du Médicament et des Produits de Santé (reference 20160072000007).
Collapse
Affiliation(s)
- Nicolas Tessandier
- CIRB, CNRS, INSERM, Collège de France, Université PSL, Paris, France
- MIVEGEC, CNRS, IRD, Université de Montpellier, France
| | - Baptiste Elie
- CIRB, CNRS, INSERM, Collège de France, Université PSL, Paris, France
- MIVEGEC, CNRS, IRD, Université de Montpellier, France
| | - Vanina Boué
- MIVEGEC, CNRS, IRD, Université de Montpellier, France
| | - Christian Selinger
- MIVEGEC, CNRS, IRD, Université de Montpellier, France
- Swiss Tropical and Public Health Institute, Basel, Switzerland
| | | | - Claire Bernat
- MIVEGEC, CNRS, IRD, Université de Montpellier, France
- CNRS UMR 5203, Institut de Génomique Fonctionnelle, Montpellier, France
| | | | - Soraya Groc
- MIVEGEC, CNRS, IRD, Université de Montpellier, France
- PCCEI, Univ. Montpellier, Inserm, EFS, Montpellier, France
| | | | | | - Marine Bonneau
- Department of Obstetrics and Gynaecology, Centre Hospitalier Universitaire de Montpellier, Montpellier, France
| | - Christelle Graf
- Department of Obstetrics and Gynaecology, Centre Hospitalier Universitaire de Montpellier, Montpellier, France
| | - Nathalie Jacobs
- Laboratory of Cellular and Molecular Immunology, GIGA Institute, University of Liège, Liège, Belgium
| | - Tsukushi Kamiya
- CIRB, CNRS, INSERM, Collège de France, Université PSL, Paris, France
| | | | - Julie Lajoie
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Canada
| | - Imène Melki
- CIRB, CNRS, INSERM, Collège de France, Université PSL, Paris, France
| | - Jean-Luc Prétet
- Université de Franche-Comté, CNRS, Chrono-environnement, Besançon, France
- Centre National de Référence Papillomavirus, CHRU de Besançon, France
| | - Bastien Reyné
- MIVEGEC, CNRS, IRD, Université de Montpellier, France
| | - Géraldine Schlecht-Louf
- UMR996, Inflammation, Chemokines and Immunopathology, INSERM, Université Paris-Saclay, Orsay, France
| | - Mircea T. Sofonea
- PCCEI, Univ. Montpellier, Inserm, EFS, Montpellier, France
- CHU de Nîmes, Nîmes, France
| | - Olivier Supplisson
- CIRB, CNRS, INSERM, Collège de France, Université PSL, Paris, France
- Sorbonne Université, France
| | - Chris Wymant
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | | | | | - Christophe Hirtz
- RMB-PPC, INM, Univ Montpellier, CHU Montpellier, INSERM CNRS, Montpellier, France
| | - Marie-Christine Picot
- Department of Medical Information (DIM), Centre Hospitalier Universitaire de Montpellier, Montpellier, France
| | - Jacques Reynes
- Department of Infectious and Tropical Diseases, Centre Hospitalier Universitaire de Montpellier, Montpellier, France
| | - Vincent Tribout
- Center for Free Information, Screening and Diagnosis (CeGIDD), Centre Hospitalier Universitaire de Montpellier, Montpellier, France
| | | | - Tim Waterboer
- German Cancer Research Center (DKFZ), Infections and Cancer Epidemiology, Heidelberg, Germany
| | - Michel Segondy
- PCCEI, Univ. Montpellier, Inserm, EFS, Montpellier, France
| | | | | | - Carmen Lía Murall
- MIVEGEC, CNRS, IRD, Université de Montpellier, France
- National Microbiology Laboratory (NML), Public Health Agency of Canada (PHAC), Canada
| | - Samuel Alizon
- CIRB, CNRS, INSERM, Collège de France, Université PSL, Paris, France
- MIVEGEC, CNRS, IRD, Université de Montpellier, France
| |
Collapse
|
33
|
Rachubinski AL, Wallace E, Gurnee E, Enriquez-Estrada BA, Worek KR, Smith KP, Araya P, Waugh KA, Granrath RE, Britton E, Lyford HR, Donovan MG, Eduthan NP, Hill AA, Martin B, Sullivan KD, Patel L, Fidler DJ, Galbraith MD, Dunnick CA, Norris DA, Espinosa JM. JAK inhibition decreases the autoimmune burden in Down syndrome. eLife 2024; 13:RP99323. [PMID: 39737640 DOI: 10.7554/elife.99323] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2025] Open
Abstract
Background Individuals with Down syndrome (DS), the genetic condition caused by trisomy 21 (T21), display clear signs of immune dysregulation, including high rates of autoimmunity and severe complications from infections. Although it is well established that T21 causes increased interferon responses and JAK/STAT signaling, elevated autoantibodies, global immune remodeling, and hypercytokinemia, the interplay between these processes, the clinical manifestations of DS, and potential therapeutic interventions remain ill defined. Methods We report a comprehensive analysis of immune dysregulation at the clinical, cellular, and molecular level in hundreds of individuals with DS, including autoantibody profiling, cytokine analysis, and deep immune mapping. We also report the interim analysis of a Phase II clinical trial investigating the safety and efficacy of the JAK inhibitor tofacitinib through multiple clinical and molecular endpoints. Results We demonstrate multi-organ autoimmunity of pediatric onset concurrent with unexpected autoantibody-phenotype associations in DS. Importantly, constitutive immune remodeling and hypercytokinemia occur from an early age prior to autoimmune diagnoses or autoantibody production. Analysis of the first 10 participants to complete 16 weeks of tofacitinib treatment shows a good safety profile and no serious adverse events. Treatment reduced skin pathology in alopecia areata, psoriasis, and atopic dermatitis, while decreasing interferon scores, cytokine scores, and levels of pathogenic autoantibodies without overt immune suppression. Conclusions JAK inhibition is a valid strategy to treat autoimmune conditions in DS. Additional research is needed to define the effects of JAK inhibition on the broader developmental and clinical hallmarks of DS. Funding NIAMS, Global Down Syndrome Foundation. Clinical trial number NCT04246372.
Collapse
Affiliation(s)
- Angela L Rachubinski
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, United States
- Department of Pediatrics, Section of Developmental Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Elizabeth Wallace
- Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Emily Gurnee
- Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Belinda A Enriquez-Estrada
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Kayleigh R Worek
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Keith P Smith
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Paula Araya
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Katherine A Waugh
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Ross E Granrath
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Eleanor Britton
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Hannah R Lyford
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Micah G Donovan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, United States
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Neetha Paul Eduthan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Amanda A Hill
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Barry Martin
- Department of Internal Medicine, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Kelly D Sullivan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, United States
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Lina Patel
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, United States
- Department of Psychiatry, Child and Adolescent Division, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Deborah J Fidler
- Department of Human Development and Family Studies, Colorado State University, Fort Collins, United States
| | - Matthew D Galbraith
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, United States
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Cory A Dunnick
- Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - David A Norris
- Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Joaquín M Espinosa
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, United States
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, United States
| |
Collapse
|
34
|
Orehek S, Ramuta TŽ, Lainšček D, Malenšek Š, Šala M, Benčina M, Jerala R, Hafner-Bratkovič I. Cytokine-armed pyroptosis induces antitumor immunity against diverse types of tumors. Nat Commun 2024; 15:10801. [PMID: 39737979 DOI: 10.1038/s41467-024-55083-3] [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: 04/12/2024] [Accepted: 11/29/2024] [Indexed: 01/01/2025] Open
Abstract
Inflammasomes are defense complexes that utilize cytokines and immunogenic cell death (ICD) to stimulate the immune system against pathogens. Inspired by their dual action, we present cytokine-armed pyroptosis as a strategy for boosting immune response against diverse types of tumors. To induce pyroptosis, we utilize designed tightly regulated gasdermin D variants comprising different pore-forming capabilities and diverse modes of activation, representing a toolbox of ICD inducers. We demonstrate that the electrogenic transfer of ICD effector-encoding plasmids into mouse melanoma tumors when combined with intratumoral expression of cytokines IL-1β, IL-12, or IL-18, enhanced anti-tumor immune responses. Careful selection of immunostimulatory molecules is, however, imperative as a combination of IL-1β and IL-18 antagonized the protective effect of pyroptosis by IFNγ-mediated upregulation of several immunosuppressive pathways. Additionally, we show that the intratumoral introduction of armed pyroptosis provides protection against distant tumors and proves effective across various tumor types without inducing systemic inflammation. Deconstructed inflammasomes thus serve as a powerful, tunable, and tumor-agnostic strategy to enhance antitumor response, even against the most resilient types of tumors.
Collapse
Affiliation(s)
- Sara Orehek
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Ljubljana, Slovenia
- Interdisciplinary Doctoral Study of Biomedicine, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Taja Železnik Ramuta
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Duško Lainšček
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Ljubljana, Slovenia
- EN-FIST Centre of Excellence, Ljubljana, Slovenia
- Centre for the Technologies of Gene and Cell Therapy, National Institute of Chemistry, Ljubljana, Slovenia
| | - Špela Malenšek
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Ljubljana, Slovenia
- Interdisciplinary Doctoral Study of Biomedicine, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Martin Šala
- Department of Analytical Chemistry, National Institute of Chemistry, Ljubljana, Slovenia
| | - Mojca Benčina
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Ljubljana, Slovenia
- Centre for the Technologies of Gene and Cell Therapy, National Institute of Chemistry, Ljubljana, Slovenia
- Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Roman Jerala
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Ljubljana, Slovenia
- EN-FIST Centre of Excellence, Ljubljana, Slovenia
- Centre for the Technologies of Gene and Cell Therapy, National Institute of Chemistry, Ljubljana, Slovenia
| | - Iva Hafner-Bratkovič
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Ljubljana, Slovenia.
- EN-FIST Centre of Excellence, Ljubljana, Slovenia.
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.
| |
Collapse
|
35
|
Lapoujade C, Blanco M, Givelet M, Gille AS, Allemand I, Lenez L, Thiounn N, Roux S, Wolf JP, Patrat C, Riou L, Barraud-Lange V, Fouchet P. Characterisation and hierarchy of the spermatogonial stem cell compartment in human spermatogenesis by spectral cytometry using a 16-colors panel. Cell Mol Life Sci 2024; 82:15. [PMID: 39725808 DOI: 10.1007/s00018-024-05496-6] [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: 04/15/2024] [Revised: 10/02/2024] [Accepted: 10/28/2024] [Indexed: 12/28/2024]
Abstract
About one in six couples experience fertility problems, and male infertility accounts for about half of these cases. Spermatogenesis originates from a small pool of spermatogonial stem cells (SSCs), which are of interest for the treatment of infertility but remain poorly characterised in humans. Using multiparametric spectral flow cytometric analysis with a 16-colours (16-C) panel of cell markers, we identify novel markers of SSCs and provide insights into unravelling and resolving the heterogeneity of the human spermatogonial cells. This 16-C panel of markers allowed the identification of a primitive SSCs state with the β2M-CD51/61-ITGA6+SSEA4+TSPAN33+THY1+CD9medEPCAMmedCD155+CD148+CD47highCD7high phenotype, with a profile close to the most primitive SSCs states 0 and SSC1-B previously defined by sc-RNAseq approach. The hierarchy of events in the spermatogonial stem cell and progenitor compartment of human spermatogenesis can be delineated. This highlights the importance of a multi-parametric and spectral cytometry approach. The in-depth characterisation of testicular cells should help to overcome the lack of stem cell knowledge, that hinders the understanding of the regenerative potential of SSCs, and is a critical parameter for the successful development of new SSCs-based cell therapies.
Collapse
Affiliation(s)
- C Lapoujade
- Université Paris-Saclay, CEA, UMR Stabilité Génétique Cellules Souches Et Radiations, iRCM/IBFJ, Laboratoire Des Cellules Souches Germinales, 92265, Fontenay-Aux-Roses, France
- Université Paris Cité, CEA, UMR Stabilité Génétique Cellules Souches Et Radiations, iRCM/IBFJ, Laboratoire Des Cellules Souches Germinales, 92265, Fontenay-Aux-Roses, France
| | - M Blanco
- Université Paris-Saclay, CEA, UMR Stabilité Génétique Cellules Souches Et Radiations, iRCM/IBFJ, Laboratoire Des Cellules Souches Germinales, 92265, Fontenay-Aux-Roses, France
- Université Paris Cité, CEA, UMR Stabilité Génétique Cellules Souches Et Radiations, iRCM/IBFJ, Laboratoire Des Cellules Souches Germinales, 92265, Fontenay-Aux-Roses, France
| | - M Givelet
- Université Paris-Saclay, CEA, UMR Stabilité Génétique Cellules Souches Et Radiations, iRCM/IBFJ, Laboratoire Des Cellules Souches Germinales, 92265, Fontenay-Aux-Roses, France
- Université Paris Cité, CEA, UMR Stabilité Génétique Cellules Souches Et Radiations, iRCM/IBFJ, Laboratoire Des Cellules Souches Germinales, 92265, Fontenay-Aux-Roses, France
- Team From Gametes To Birth. Departments Genetic and Cellular Plasticity. Metabolism and Endocrinology. Cochin Institute. INSERM U1016, Paris, France
- Université Paris Cité, Paris, France
- AP-HP. Center-University Paris Cité. Cochin Hospital, Paris, France
| | - A S Gille
- Université Paris-Saclay, CEA, UMR Stabilité Génétique Cellules Souches Et Radiations, iRCM/IBFJ, Laboratoire Des Cellules Souches Germinales, 92265, Fontenay-Aux-Roses, France
- Université Paris Cité, CEA, UMR Stabilité Génétique Cellules Souches Et Radiations, iRCM/IBFJ, Laboratoire Des Cellules Souches Germinales, 92265, Fontenay-Aux-Roses, France
- Team From Gametes To Birth. Departments Genetic and Cellular Plasticity. Metabolism and Endocrinology. Cochin Institute. INSERM U1016, Paris, France
- Université Paris Cité, Paris, France
- AP-HP. Center-University Paris Cité. Cochin Hospital, Paris, France
| | - I Allemand
- Université Paris-Saclay, CEA, UMR Stabilité Génétique Cellules Souches Et Radiations, iRCM/IBFJ, Laboratoire Des Cellules Souches Germinales, 92265, Fontenay-Aux-Roses, France
- Université Paris Cité, CEA, UMR Stabilité Génétique Cellules Souches Et Radiations, iRCM/IBFJ, Laboratoire Des Cellules Souches Germinales, 92265, Fontenay-Aux-Roses, France
| | - L Lenez
- Team From Gametes To Birth. Departments Genetic and Cellular Plasticity. Metabolism and Endocrinology. Cochin Institute. INSERM U1016, Paris, France
| | - N Thiounn
- AP-HP. Center-University Paris Cité. Cochin Hospital, Paris, France
| | - S Roux
- AP-HP. Center-University Paris Cité. Cochin Hospital, Paris, France
| | - J P Wolf
- Team From Gametes To Birth. Departments Genetic and Cellular Plasticity. Metabolism and Endocrinology. Cochin Institute. INSERM U1016, Paris, France
- Université Paris Cité, Paris, France
- AP-HP. Center-University Paris Cité. Cochin Hospital, Paris, France
| | - C Patrat
- Team From Gametes To Birth. Departments Genetic and Cellular Plasticity. Metabolism and Endocrinology. Cochin Institute. INSERM U1016, Paris, France
- Université Paris Cité, Paris, France
- AP-HP. Center-University Paris Cité. Cochin Hospital, Paris, France
| | - L Riou
- Université Paris-Saclay, CEA, UMR Stabilité Génétique Cellules Souches Et Radiations, iRCM/IBFJ, Laboratoire Des Cellules Souches Germinales, 92265, Fontenay-Aux-Roses, France
- Université Paris Cité, CEA, UMR Stabilité Génétique Cellules Souches Et Radiations, iRCM/IBFJ, Laboratoire Des Cellules Souches Germinales, 92265, Fontenay-Aux-Roses, France
| | - V Barraud-Lange
- Team From Gametes To Birth. Departments Genetic and Cellular Plasticity. Metabolism and Endocrinology. Cochin Institute. INSERM U1016, Paris, France
- Université Paris Cité, Paris, France
- AP-HP. Center-University Paris Cité. Cochin Hospital, Paris, France
| | - P Fouchet
- Université Paris-Saclay, CEA, UMR Stabilité Génétique Cellules Souches Et Radiations, iRCM/IBFJ, Laboratoire Des Cellules Souches Germinales, 92265, Fontenay-Aux-Roses, France.
- Université Paris Cité, CEA, UMR Stabilité Génétique Cellules Souches Et Radiations, iRCM/IBFJ, Laboratoire Des Cellules Souches Germinales, 92265, Fontenay-Aux-Roses, France.
| |
Collapse
|
36
|
Capelletti MM, Montini O, Ruini E, Tettamanti S, Savino AM, Sarno J. Unlocking the Heterogeneity in Acute Leukaemia: Dissection of Clonal Architecture and Metabolic Properties for Clinical Interventions. Int J Mol Sci 2024; 26:45. [PMID: 39795903 PMCID: PMC11719665 DOI: 10.3390/ijms26010045] [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: 11/21/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 01/13/2025] Open
Abstract
Genetic studies of haematological cancers have pointed out the heterogeneity of leukaemia in its different subpopulations, with distinct mutations and characteristics, impacting the treatment response. Next-generation sequencing (NGS) and genome-wide analyses, as well as single-cell technologies, have offered unprecedented insights into the clonal heterogeneity within the same tumour. A key component of this heterogeneity that remains unexplored is the intracellular metabolome, a dynamic network that determines cell functions, signalling, epigenome regulation, immunity and inflammation. Understanding the metabolic diversities among cancer cells and their surrounding environments is therefore essential in unravelling the complexities of leukaemia and improving therapeutic strategies. Here, we describe the currently available methodologies and approaches to addressing the dynamic heterogeneity of leukaemia progression. In the second section, we focus on metabolic leukaemic vulnerabilities in acute myeloid leukaemia (AML) and acute lymphoblastic leukaemia (ALL). Lastly, we provide a comprehensive overview of the most interesting clinical trials designed to target these metabolic dependencies, highlighting their potential to advance therapeutic strategies in leukaemia treatment. The integration of multi-omics data for cancer identification with the metabolic states of tumour cells will enable a comprehensive "micro-to-macro" approach for the refinement of clinical practices and delivery of personalised therapies.
Collapse
Affiliation(s)
- Martina Maria Capelletti
- School of Medicine and Surgery, University of Milan-Bicocca, 20126 Milan, Italy; (M.M.C.); (O.M.); (E.R.); (A.M.S.)
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Orsola Montini
- School of Medicine and Surgery, University of Milan-Bicocca, 20126 Milan, Italy; (M.M.C.); (O.M.); (E.R.); (A.M.S.)
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Emilio Ruini
- School of Medicine and Surgery, University of Milan-Bicocca, 20126 Milan, Italy; (M.M.C.); (O.M.); (E.R.); (A.M.S.)
| | - Sarah Tettamanti
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Angela Maria Savino
- School of Medicine and Surgery, University of Milan-Bicocca, 20126 Milan, Italy; (M.M.C.); (O.M.); (E.R.); (A.M.S.)
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Jolanda Sarno
- School of Medicine and Surgery, University of Milan-Bicocca, 20126 Milan, Italy; (M.M.C.); (O.M.); (E.R.); (A.M.S.)
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| |
Collapse
|
37
|
Luscombe C, Jones E, Gregorova M, Jones N, Rivino L. Impact of cryopreservation on immune cell metabolism as measured by SCENITH. OXFORD OPEN IMMUNOLOGY 2024; 6:iqae015. [PMID: 39906176 PMCID: PMC11790226 DOI: 10.1093/oxfimm/iqae015] [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: 05/27/2024] [Revised: 09/29/2024] [Accepted: 12/04/2024] [Indexed: 02/06/2025] Open
Abstract
The dynamic functioning of immune cells is regulated by cellular metabolic processes, and there is growing interest in the study of immunometabolic correlates of dysfunctional immune responses. SCENITH is a novel flow cytometry-based technique that allows for ex vivo metabolic profiling of immune cells within heterogeneous samples. Cryopreservation of clinical samples is frequently undertaken to facilitate high throughput processing and longitudinal analyses of immune responses, but is thought to lead to cellular metabolic dysfunction. We aimed to investigate the impact of cryopreservation on immune cell metabolism, harnessing SCENITH's unique ability to describe the divergent bioenergetic characteristics of distinct immune cell subsets. We demonstrate that upon activation, T cells are unable to sufficiently/readily undergo metabolic reprogramming. Additionally, we find that cryopreservation introduces a time-dependent metabolic artefact that favours glycolysis and impairs oxidative phosphorylation, suggesting that cryopreservation results in mitochondrial dysfunction. Despite this artefact, SCENITH was still able to reveal the distinct bioenergetic profiles of contrasting immune cells populations following cryopreservation. Whilst SCENITH can provide valuable information about immune cell metabolism even in cryopreserved samples, our findings have important implications for the design of future studies. Investigators should carefully consider how to process and store clinical samples to ensure that cryopreservation does not confound analyses, particularly where longitudinal sampling is required.
Collapse
Affiliation(s)
- Curtis Luscombe
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, United Kingdom
| | - Eben Jones
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, United Kingdom
| | - Michaela Gregorova
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, United Kingdom
| | - Nicholas Jones
- Institute of Life Science, Swansea University Medical School, Swansea University, SA2 8PP, United Kingdom
| | - Laura Rivino
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, United Kingdom
| |
Collapse
|
38
|
Mocking TR, Kelder A, Reuvekamp T, Ngai LL, Rutten P, Gradowska P, van de Loosdrecht AA, Cloos J, Bachas C. Computational assessment of measurable residual disease in acute myeloid leukemia using mixture models. COMMUNICATIONS MEDICINE 2024; 4:271. [PMID: 39702555 DOI: 10.1038/s43856-024-00700-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 12/05/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND The proportion of residual leukemic blasts after chemotherapy assessed by multiparameter flow cytometry, is an important prognostic factor for the risk of relapse and overall survival in acute myeloid leukemia (AML). This measurable residual disease (MRD) is used in clinical trials to stratify patients for more or less intensive consolidation therapy. However, an objective and reproducible analysis method to assess MRD status from flow cytometry data is lacking, yet is highly anticipated for broader implementation of MRD testing. METHODS We propose a computational pipeline based on Gaussian mixture modeling that allows a fully automated assessment of MRD status while remaining completely interpretable for clinical diagnostic experts. Our pipeline requires limited training data, which makes it easily transferable to other medical centers and cytometry platforms. RESULTS We identify all healthy and leukemic immature myeloid cells in with high concordance (Spearman's Rho = 0.974) and classification performance (median F-score = 0.861) compared to manual analysis. Using control samples (n = 18), we calculate a computational MRD percentage with high concordance to expert gating (Spearman's rho = 0.823) and predict MRD status in a cohort of 35 AML follow-up measurements with high accuracy (97%). CONCLUSIONS We demonstrate that our pipeline provides a powerful tool for fast (~3 s) and objective automated MRD assessment in AML.
Collapse
Affiliation(s)
- Tim R Mocking
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Angèle Kelder
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Tom Reuvekamp
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Department of Hematology, Amsterdam UMC, Universiteit van Amsterdam, Amsterdam, The Netherlands
| | - Lok Lam Ngai
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Philip Rutten
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Department of Epidemiology and Data Science, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Patrycja Gradowska
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
- HOVON Foundation, Rotterdam, The Netherlands
| | - Arjan A van de Loosdrecht
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Jacqueline Cloos
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Costa Bachas
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands.
| |
Collapse
|
39
|
Kröger C, Müller S, Leidner J, Kröber T, Warnat-Herresthal S, Spintge JB, Zajac T, Neubauer A, Frolov A, Carraro C, Jessen F, Puccio S, Aschenbrenner AC, Schultze JL, Pecht T, Beyer MD, Bonaguro L. Unveiling the power of high-dimensional cytometry data with cyCONDOR. Nat Commun 2024; 15:10702. [PMID: 39702306 DOI: 10.1038/s41467-024-55179-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 11/26/2024] [Indexed: 12/21/2024] Open
Abstract
High-dimensional cytometry (HDC) is a powerful technology for studying single-cell phenotypes in complex biological systems. Although technological developments and affordability have made HDC broadly available in recent years, technological advances were not coupled with an adequate development of analytical methods that can take full advantage of the complex data generated. While several analytical platforms and bioinformatics tools have become available for the analysis of HDC data, these are either web-hosted with limited scalability or designed for expert computational biologists, making their use unapproachable for wet lab scientists. Additionally, end-to-end HDC data analysis is further hampered due to missing unified analytical ecosystems, requiring researchers to navigate multiple platforms and software packages to complete the analysis. To bridge this data analysis gap in HDC we develop cyCONDOR, an easy-to-use computational framework covering not only all essential steps of cytometry data analysis but also including an array of downstream functions and tools to expand the biological interpretation of the data. The comprehensive suite of features of cyCONDOR, including guided pre-processing, clustering, dimensionality reduction, and machine learning algorithms, facilitates the seamless integration of cyCONDOR into clinically relevant settings, where scalability and disease classification are paramount for the widespread adoption of HDC in clinical practice. Additionally, the advanced analytical features of cyCONDOR, such as pseudotime analysis and batch integration, provide researchers with the tools to extract deeper insights from their data. We use cyCONDOR on a variety of data from different tissues and technologies demonstrating its versatility to assist the analysis of high-dimensional data from preprocessing to biological interpretation.
Collapse
Affiliation(s)
- Charlotte Kröger
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Genomics & Immunoregulation, LIMES Institute, University of Bonn, Bonn, Germany
| | - Sophie Müller
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Genomics & Immunoregulation, LIMES Institute, University of Bonn, Bonn, Germany
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Jacqueline Leidner
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Genomics & Immunoregulation, LIMES Institute, University of Bonn, Bonn, Germany
| | - Theresa Kröber
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Stefanie Warnat-Herresthal
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Genomics & Immunoregulation, LIMES Institute, University of Bonn, Bonn, Germany
| | - Jannis Bastian Spintge
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics, DZNE and University of Bonn and West German Genome Center, Bonn, Germany
| | - Timo Zajac
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Anna Neubauer
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Aleksej Frolov
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Immunogenomics & Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Caterina Carraro
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Genomics & Immunoregulation, LIMES Institute, University of Bonn, Bonn, Germany
| | - Frank Jessen
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Venusberg-Campus 1, Bonn, Germany
- Department of Psychiatry, University of Cologne, Medical Faculty, Kerpener Strasse 62, Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Strasse 26, Köln, Germany
| | - Simone Puccio
- Laboratory of Translational Immunology, IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, Milan, Italy
- Institute of Genetic and Biomedical Research, UoS Milan, National Research Council, via Manzoni 56, Rozzano, Milan, Italy
| | - Anna C Aschenbrenner
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Joachim L Schultze
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Genomics & Immunoregulation, LIMES Institute, University of Bonn, Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics, DZNE and University of Bonn and West German Genome Center, Bonn, Germany
| | - Tal Pecht
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Genomics & Immunoregulation, LIMES Institute, University of Bonn, Bonn, Germany
| | - Marc D Beyer
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics, DZNE and University of Bonn and West German Genome Center, Bonn, Germany
- Immunogenomics & Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Lorenzo Bonaguro
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.
- Genomics & Immunoregulation, LIMES Institute, University of Bonn, Bonn, Germany.
| |
Collapse
|
40
|
Wang YW, Tan PC, Li QF, Xu XW, Zhou SB. Adipose tissue protects against skin photodamage through CD151- and AdipoQ- EVs. Cell Commun Signal 2024; 22:594. [PMID: 39696450 DOI: 10.1186/s12964-024-01978-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024] Open
Abstract
To clarify the protective effects of subcutaneous adipose tissue (SAT) against photodamage, we utilized nude mouse skin with or without SAT. Skin and fibroblasts were treated with adipose tissue-derived extracellular vesicles (AT-EVs) or extracellular vesicles derived from adipose-derived stem cells (ADSC-EVs) to demonstrate that SAT protects the overlying skin from photodamage primarily through AT-EVs. Surprisingly, AT-EVs stimulated fibroblast proliferation more rapidly than ADSC-EVs did. The yield of AT-EVs from the same volume of AT was 200 times greater than that of ADSC-EVs. To compare the differences between AT-EVs and ADSC-EVs, we used a proximity barcoding assay (PBA) to analyze the surface proteins on individual particles of these two types of EVs. PBA analysis revealed that AT-EVs contain diverse subpopulations, with 83.42% expressing CD151, compared to only 1.98% of ADSC-EVs. Furthermore, AT-EVs are internalized more rapidly by cells than ADSC-EVs, as our study demonstrated that CD151-positive AT-EVs were endocytosed more quickly than their CD151-negative counterparts. Additionally, adiponectin in AT-EVs activated the AMPK pathway and inhibited the NF-κB pathway, enhancing fibroblast protection against photodamage. The significantly higher yield and faster acquisition of AT-EVs compared to ADSC-EVs underscore their potential for broader applications.
Collapse
Affiliation(s)
- Yan-Wen Wang
- Department of Plastic & Reconstructive Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Poh-Ching Tan
- Department of Plastic & Reconstructive Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qing-Feng Li
- Department of Plastic & Reconstructive Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xue-Wen Xu
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, China.
| | - Shuang-Bai Zhou
- Department of Plastic & Reconstructive Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
41
|
Sun H, Yu S, Casals AM, Bäckström A, Lu Y, Lindskog C, Ruffalo M, Lundberg E, Murphy RF. Flexible and robust cell type annotation for highly multiplexed tissue images. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.12.612510. [PMID: 39345395 PMCID: PMC11429614 DOI: 10.1101/2024.09.12.612510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Identifying cell types in highly multiplexed images is essential for understanding tissue spatial organization. Current cell type annotation methods often rely on extensive reference images and manual adjustments. In this work, we present a tool, Robust Image-Based Cell Annotator (RIBCA), that enables accurate, automated, unbiased, and fine-grained cell type annotation for images with a wide range of antibody panels, without requiring additional model training or human intervention. Our tool has successfully annotated over 3 million cells, revealing the spatial organization of various cell types across more than 40 different human tissues. It is open-source and features a modular design, allowing for easy extension to additional cell types.
Collapse
Affiliation(s)
- Huangqingbo Sun
- Ray and Stephanie Lane Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Shiqiu Yu
- Ray and Stephanie Lane Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA, USA
| | | | - Anna Bäckström
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Yuxin Lu
- Ray and Stephanie Lane Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Cecilia Lindskog
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Matthew Ruffalo
- Ray and Stephanie Lane Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Emma Lundberg
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH-Royal Institute of Technology, Stockholm, Sweden
- Department of Pathology, Stanford University, Stanford, CA, USA
- Chan Zuckerberg Biohub San Francisco, San Francisco, CA, USA
| | - Robert F Murphy
- Ray and Stephanie Lane Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA, USA
| |
Collapse
|
42
|
Kuijper LH, Kreher C, Elias G, Claireaux M, Kerster G, Bos AV, Duurland MC, Konijn VAL, Paul AGA, de Jong N, de Jongh R, Steenhuis M, Garcia-Vallejo JJ, van Gils MJ, Kuijpers TW, Eftimov F, Rispens T, van der Schoot CE, van Ham SM, ten Brinke A. Longevity of antibody responses is associated with distinct antigen-specific B cell subsets early after infection. Front Immunol 2024; 15:1505719. [PMID: 39742271 PMCID: PMC11686410 DOI: 10.3389/fimmu.2024.1505719] [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: 10/03/2024] [Accepted: 11/12/2024] [Indexed: 01/03/2025] Open
Abstract
Introduction Upon infection, T cell-driven B cell responses in GC reactions induce memory B cells and antibody-secreting cells that secrete protective antibodies. How formation of specifically long-lived plasma cells is regulated via the interplay between specific B and CD4+ T cells is not well understood. Generally, antibody levels decline over time after clearance of the primary infection. Method In this study, convalescent individuals with stable RBD antibody levels (n=14, "sustainers") were compared with donors (n=13) with the greatest antibody decline from a cohort of 132. To investigate the role of the cellular immune compartment in the maintenance of antibody levels, SARS-CoV-2-specific responses at 4 to 6 weeks post-mild COVID-19 infection were characterized using deep immune profiling. Results Both groups had similar frequencies of total SARS-CoV-2-specific B and CD4+ T cells. Sustainers had fewer Spike-specific IgG+ memory B cells early after infection and increased neutralizing capacity of RBD antibodies over time, unlike the declining group. However, declining IgG titers correlated with lower frequency of Spike-specific CD4+ T cells. Conclusion These data suggest that "sustainers" have unique dynamics of GC reactions, yield different outputs of terminally differentiating cells, and improve the quality of protective antibodies over time. This study helps identify factors controlling formation of long-lived PC and sustained antibody responses.
Collapse
Affiliation(s)
- Lisan H. Kuijper
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Christine Kreher
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - George Elias
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Mathieu Claireaux
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, Netherlands
- Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
| | - Gius Kerster
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, Netherlands
- Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
| | - Amélie V. Bos
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Mariël C. Duurland
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Veronique A. L. Konijn
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Alberta G. A. Paul
- Cytek Biosciences, Inc., Fremont, CA, United States
- Department of Molecular Cell Biology and Immunology, Amsterdam Infection & Immunity and Cancer Center Amsterdam, Amsterdam University Medical Centers, Free University of Amsterdam, Amsterdam, Netherlands
| | - Nina de Jong
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Rivka de Jongh
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Maurice Steenhuis
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Juan J. Garcia-Vallejo
- Department of Molecular Cell Biology and Immunology, Amsterdam Infection & Immunity and Cancer Center Amsterdam, Amsterdam University Medical Centers, Free University of Amsterdam, Amsterdam, Netherlands
| | - Marit J. van Gils
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, Netherlands
- Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam UMC Location University of Amsterdam, Amsterdam, Netherlands
| | - Taco W. Kuijpers
- Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma Children’s Hospital, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Filip Eftimov
- Department of Neurology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Theo Rispens
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - C. Ellen van der Schoot
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - S. Marieke van Ham
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Anja ten Brinke
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
43
|
Siam MHB, Ali MA, Vardaman D, Acharyya S, Patil M, Tyrrell DJ. CAFE: An Integrated Web App for High-Dimensional Analysis and Visualization in Spectral Flow Cytometry. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.03.626714. [PMID: 39713432 PMCID: PMC11661093 DOI: 10.1101/2024.12.03.626714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Spectral flow cytometry provides greater insights into cellular heterogeneity by simultaneous measurement of up to 50 markers. However, analyzing such high-dimensional (HD) data is complex through traditional manual gating strategy. To address this gap, we developed CAFE as an open-source Python-based web application with a graphical user interface. Built with Streamlit, CAFE incorporates libraries such as Scanpy for single-cell analysis, Pandas and PyArrow for efficient data handling, and Matplotlib, Seaborn, Plotly for creating customizable figures. Its robust toolset includes density-based down-sampling, dimensionality reduction, batch correction, Leiden-based clustering, cluster merging and annotation. Using CAFE, we demonstrated analysis of a human PBMC dataset of 350,000 cells identifying 16 distinct cell clusters. CAFE can generate publication-ready figures in real time via interactive slider controls and dropdown menus, eliminating the need for coding expertise and making HD data analysis accessible to all. CAFE is licensed under MIT and is freely available at https://github.com/mhbsiam/cafe.
Collapse
Affiliation(s)
- Md Hasanul Banna Siam
- Department of Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35205 USA
| | - Md Akkas Ali
- Department of Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35205 USA
| | - Donald Vardaman
- Department of Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35205 USA
| | - Satwik Acharyya
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL, 35233 USA
- Department of Biomedical Informatics and Data Science, University of Alabama at Birmingham, Birmingham, AL, 35294 USA
| | - Mallikarjun Patil
- Department of Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35205 USA
| | - Daniel J. Tyrrell
- Department of Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35205 USA
| |
Collapse
|
44
|
Mistri SK, Hilton BM, Horrigan KJ, Andretta ES, Savard R, Dienz O, Hampel KJ, Gerrard DL, Rose JT, Sidiropoulos N, Majumdar D, Boyson JE. SLAM/SAP signaling regulates discrete γδ T cell developmental checkpoints and shapes the innate-like γδ TCR repertoire. eLife 2024; 13:RP97229. [PMID: 39656519 PMCID: PMC11630817 DOI: 10.7554/elife.97229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2024] Open
Abstract
During thymic development, most γδ T cells acquire innate-like characteristics that are critical for their function in tumor surveillance, infectious disease, and tissue repair. The mechanisms, however, that regulate γδ T cell developmental programming remain unclear. Recently, we demonstrated that the SLAM/SAP signaling pathway regulates the development and function of multiple innate-like γδ T cell subsets. Here, we used a single-cell proteogenomics approach to identify SAP-dependent developmental checkpoints and to define the SAP-dependent γδ TCR repertoire in mice. SAP deficiency resulted in both a significant loss of an immature Gzma+Blk+Etv5+Tox2+ γδT17 precursor population and a significant increase in Cd4+Cd8+Rorc+Ptcra+Rag1+ thymic γδ T cells. SAP-dependent diversion of embryonic day 17 thymic γδ T cell clonotypes into the αβ T cell developmental pathway was associated with a decreased frequency of mature clonotypes in neonatal thymus, and an altered γδ TCR repertoire in the periphery. Finally, we identify TRGV4/TRAV13-4(DV7)-expressing T cells as a novel, SAP-dependent Vγ4 γδT1 subset. Together, the data support a model in which SAP-dependent γδ/αβ T cell lineage commitment regulates γδ T cell developmental programming and shapes the γδ TCR repertoire.
Collapse
MESH Headings
- Animals
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Receptors, Antigen, T-Cell, gamma-delta/genetics
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Mice
- Signal Transduction
- Signaling Lymphocytic Activation Molecule Associated Protein/metabolism
- Signaling Lymphocytic Activation Molecule Associated Protein/genetics
- Immunity, Innate
- Mice, Inbred C57BL
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- Thymus Gland/immunology
- Thymus Gland/metabolism
- Cell Differentiation
- Intraepithelial Lymphocytes/immunology
- Intraepithelial Lymphocytes/metabolism
- Signaling Lymphocytic Activation Molecule Family
Collapse
Affiliation(s)
- Somen K Mistri
- Department of Surgery, Larner College of Medicine, University of VermontBurlingtonUnited States
| | - Brianna M Hilton
- Department of Surgery, Larner College of Medicine, University of VermontBurlingtonUnited States
| | - Katherine J Horrigan
- Department of Surgery, Larner College of Medicine, University of VermontBurlingtonUnited States
| | - Emma S Andretta
- Department of Surgery, Larner College of Medicine, University of VermontBurlingtonUnited States
| | - Remi Savard
- Department of Surgery, Larner College of Medicine, University of VermontBurlingtonUnited States
| | - Oliver Dienz
- Department of Surgery, Larner College of Medicine, University of VermontBurlingtonUnited States
| | - Kenneth J Hampel
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont Medical CenterBurlingtonUnited States
| | - Diana L Gerrard
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont Medical CenterBurlingtonUnited States
| | - Joshua T Rose
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont Medical CenterBurlingtonUnited States
| | - Nikoletta Sidiropoulos
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont Medical CenterBurlingtonUnited States
| | - Dev Majumdar
- Department of Surgery, Larner College of Medicine, University of VermontBurlingtonUnited States
| | - Jonathan E Boyson
- Department of Surgery, Larner College of Medicine, University of VermontBurlingtonUnited States
| |
Collapse
|
45
|
Coyle C, Ma M, Abraham Y, Mahony CB, Steel K, Simpson C, Guerra N, Croft AP, Rapecki S, Cope A, Bowcutt R, Perucha E. NK cell subsets define sustained remission in rheumatoid arthritis. JCI Insight 2024; 9:e182390. [PMID: 39418106 PMCID: PMC11623943 DOI: 10.1172/jci.insight.182390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024] Open
Abstract
Rheumatoid arthritis (RA) is an immune-mediated, chronic inflammatory condition. With modern therapeutics and evidence-based management strategies, achieving sustained remission is increasingly common. To prevent complications associated with prolonged use of immunosuppressants, drug tapering or withdrawal is recommended. However, due to the lack of tools that define immunological remission, disease flares are frequent, highlighting the need for a more precision medicine-based approach. Utilizing high-dimensional phenotyping platforms, we set out to define peripheral blood immunological signatures of sustained remission in RA. We identified that CD8+CD57+KIR2DL1+ NK cells are associated with sustained remission. Functional studies uncovered an NK cell subset characterized by normal degranulation responses and reduced proinflammatory cytokine expression, which was elevated in sustained remission. Furthermore, flow cytometric analysis of NK cells from synovial fluid combined with interrogation of a publicly available single-cell RNA-Seq dataset of synovial tissue from active RA identified a deficiency of the phenotypic characteristics associated with this NK cell remission signature. In summary, we have uncovered an immune signature of RA remission associated with compositional changes in NK cell phenotype and function that has implications for understanding the effect of sustained remission on host immunity and distinct features that may define operational tolerance in RA.
Collapse
Affiliation(s)
- Carl Coyle
- Centre for Inflammation Biology and Cancer Immunology, Floor 1, New Hunt’s House, Great Maze Pond, King’s College London, Guy’s Campus, London, United Kingdom
- Centre for Rheumatic Diseases, King’s College London, London, United Kingdom
| | - Margaret Ma
- Centre for Rheumatic Diseases, King’s College London, London, United Kingdom
- Level 10, Tower Block, Division of Rheumatology, University Medicine Cluster, National University Health System, Singapore
- Department of Medicine, National University Singapore, Singapore
| | | | - Christopher B. Mahony
- Rheumatology Research Group, Institute of Inflammation and Ageing, Queen Elizabeth Hospital, and
- Birmingham NIHR Biomedical Research Centre, University of Birmingham, Birmingham, United Kingdom
| | - Kathryn Steel
- Centre for Inflammation Biology and Cancer Immunology, Floor 1, New Hunt’s House, Great Maze Pond, King’s College London, Guy’s Campus, London, United Kingdom
- Centre for Rheumatic Diseases, King’s College London, London, United Kingdom
| | | | - Nadia Guerra
- Faculty of Natural Sciences, Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Adam P. Croft
- Rheumatology Research Group, Institute of Inflammation and Ageing, Queen Elizabeth Hospital, and
- Birmingham NIHR Biomedical Research Centre, University of Birmingham, Birmingham, United Kingdom
| | | | - Andrew Cope
- Centre for Inflammation Biology and Cancer Immunology, Floor 1, New Hunt’s House, Great Maze Pond, King’s College London, Guy’s Campus, London, United Kingdom
- Centre for Rheumatic Diseases, King’s College London, London, United Kingdom
| | | | - Esperanza Perucha
- Centre for Inflammation Biology and Cancer Immunology, Floor 1, New Hunt’s House, Great Maze Pond, King’s College London, Guy’s Campus, London, United Kingdom
- Centre for Rheumatic Diseases, King’s College London, London, United Kingdom
| |
Collapse
|
46
|
Vardaman D, Ali MA, Siam MHB, Bolding C, Tidwell H, Stephens H, Patil M, Tyrrell DJ. Development of a Spectral Flow Cytometry Analysis Pipeline for High-dimensional Immune Cell Characterization. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1713-1724. [PMID: 39451039 PMCID: PMC11573633 DOI: 10.4049/jimmunol.2400370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 10/02/2024] [Indexed: 10/26/2024]
Abstract
Flow cytometry is used for immune cell analysis for cell composition and function. Spectral flow cytometry allows for high-dimensional analysis of immune cells, overcoming limitations of conventional flow cytometry. However, analyzing data from large Ab panels is challenging using traditional biaxial gating strategies. We present, to our knowledge, a novel analysis pipeline to improve analysis of spectral flow cytometry. We employ this method to identify rare T cell populations in aging. We isolated splenocytes from young (2-3 mo old) and aged (18-19 mo old) female C57BL/6N mice and then stained these with a panel of 20 fluorescently labeled Abs. We performed spectral flow cytometry and then data processing and analysis using Python within a Jupyter Notebook environment to perform dimensionality reduction, batch correction, unsupervised clustering, and differential expression analysis. Our analysis of 3,776,804 T cells from 11 spleens revealed 35 distinct T cell clusters identified by surface marker expression. We observed significant differences between young and aged mice, with clusters enriched in one age group over the other. Naive, effector memory, and central memory CD8+ and CD4+ T cell subsets exhibited age-associated changes in abundance and marker expression. We also demonstrate the utility of our pipeline in a human PBMC dataset that used a 50-fluorescent color panel. By leveraging high-dimensional analysis methods, we provide insights into the immune aging process. This approach offers a robust and easily implemented analysis pipeline for spectral flow cytometry data that may facilitate the discovery of novel therapeutic targets for age-related immune dysfunction.
Collapse
Affiliation(s)
- Donald Vardaman
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35205 USA
| | - Md Akkas Ali
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35205 USA
- Biochemistry and Structural Biology Theme, Graduate Biomedical Sciences, University of Alabama at Birmingham, Birmingham, AL, 35205 USA
| | - Md Hasanul Banna Siam
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35205 USA
- Microbiology Theme, Graduate Biomedical Sciences, University of Alabama at Birmingham, Birmingham, AL, 35205 USA
| | - Chase Bolding
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35205 USA
| | - Harrison Tidwell
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35205 USA
| | - Holly Stephens
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, 35205 USA
- Immunology Theme, Graduate Biomedical Sciences, University of Alabama at Birmingham, Birmingham, AL, 35205 USA
| | - Mallikarjun Patil
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35205 USA
| | - Daniel J. Tyrrell
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35205 USA
| |
Collapse
|
47
|
Wlosik J, Granjeaud S, Gorvel L, Olive D, Chretien AS. A beginner's guide to supervised analysis for mass cytometry data in cancer biology. Cytometry A 2024; 105:853-869. [PMID: 39486897 DOI: 10.1002/cyto.a.24901] [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: 06/10/2024] [Revised: 09/16/2024] [Accepted: 10/01/2024] [Indexed: 11/04/2024]
Abstract
Mass cytometry enables deep profiling of biological samples at single-cell resolution. This technology is more than relevant in cancer research due to high cellular heterogeneity and complexity. Downstream analysis of high-dimensional datasets increasingly relies on machine learning (ML) to extract clinically relevant information, including supervised algorithms for classification and regression purposes. In cancer research, they are used to develop predictive models that will guide clinical decision making. However, the development of supervised algorithms faces major challenges, such as sufficient validation, before being translated into the clinics. In this work, we provide a framework for the analysis of mass cytometry data with a specific focus on supervised algorithms and practical examples of their applications. We also raise awareness on key issues regarding good practices for researchers curious to implement supervised ML on their mass cytometry data. Finally, we discuss the challenges of supervised ML application to cancer research.
Collapse
Affiliation(s)
- Julia Wlosik
- Team 'Immunity and Cancer', Marseille Cancer Research Center, Inserm U1068, CNRS UMR7258, Paoli-Calmettes Institute, Aix-Marseille University UM105, Marseille, France
- Immunomonitoring Department, Paoli-Calmettes Institute, Marseille, France
| | - Samuel Granjeaud
- Systems Biology Platform, Marseille Cancer Research Center, Inserm U1068, CNRS UMR7258, Paoli-Calmettes Institute, Aix-Marseille University UM105, Marseille, France
| | - Laurent Gorvel
- Team 'Immunity and Cancer', Marseille Cancer Research Center, Inserm U1068, CNRS UMR7258, Paoli-Calmettes Institute, Aix-Marseille University UM105, Marseille, France
- Immunomonitoring Department, Paoli-Calmettes Institute, Marseille, France
| | - Daniel Olive
- Team 'Immunity and Cancer', Marseille Cancer Research Center, Inserm U1068, CNRS UMR7258, Paoli-Calmettes Institute, Aix-Marseille University UM105, Marseille, France
- Immunomonitoring Department, Paoli-Calmettes Institute, Marseille, France
| | - Anne-Sophie Chretien
- Team 'Immunity and Cancer', Marseille Cancer Research Center, Inserm U1068, CNRS UMR7258, Paoli-Calmettes Institute, Aix-Marseille University UM105, Marseille, France
- Immunomonitoring Department, Paoli-Calmettes Institute, Marseille, France
| |
Collapse
|
48
|
Musial SC, Kleist SA, Degefu HN, Ford MA, Chen T, Isaacs JF, Boussiotis VA, Skorput AGJ, Rosato PC. Alarm Functions of PD-1+ Brain-Resident Memory T Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1585-1594. [PMID: 39413000 DOI: 10.4049/jimmunol.2400295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 09/27/2024] [Indexed: 10/18/2024]
Abstract
Resident memory T cells (TRM cells) have been described in barrier tissues as having a "sensing and alarm" function where, upon sensing cognate Ag, they alarm the surrounding tissue and orchestrate local recruitment and activation of immune cells. In the immunologically unique and tightly restricted CNS, it remains unclear whether and how brain TRM cells, which express the inhibitory receptor programmed cell death protein 1 (PD-1), alarm the surrounding tissue during Ag re-encounter. Using mouse models, we reveal that TRM cells are sufficient to drive the rapid remodeling of the brain immune landscape through activation of microglia, dendritic cells, NK cells, and B cells, expansion of regulatory T cells, and recruitment of macrophages and monocytic dendritic cells. Moreover, we report that although PD-1 restrained granzyme B upregulation in brain TRM cells reactivated via viral peptide, we observed no apparent effect on cytotoxicity in vivo, or downstream alarm responses within 48 h of TRM reactivation. We conclude that TRM cells are sufficient to trigger rapid immune activation and recruitment in the CNS and may have an unappreciated role in driving neuroinflammation.
Collapse
Affiliation(s)
- Shawn C Musial
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth College, Lebanon, NH
| | - Sierra A Kleist
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth College, Lebanon, NH
| | - Hanna N Degefu
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth College, Lebanon, NH
| | - Myles A Ford
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth College, Lebanon, NH
| | - Tiffany Chen
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth College, Lebanon, NH
| | - Jordan F Isaacs
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth College, Lebanon, NH
| | - Vassiliki A Boussiotis
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | | | - Pamela C Rosato
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth College, Lebanon, NH
| |
Collapse
|
49
|
Holm M, Stepanauskaitė L, Bäckström A, Birgersson M, Socciarelli F, Archer A, Stadler C, Williams C. Spatial profiling of the mouse colonic immune landscape associated with colitis and sex. Commun Biol 2024; 7:1595. [PMID: 39613949 DOI: 10.1038/s42003-024-07276-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 11/15/2024] [Indexed: 12/01/2024] Open
Abstract
Inflammatory intestinal conditions are a major disease burden. Numerous factors shape the distribution of immune cells in the colon, but a spatial characterization of the homeostatic and inflamed colonic immune microenvironment is lacking. Here, we use the COMET platform for multiplex immunofluorescence to profile the infiltration of nine immune cell populations in mice of both sexes (N = 16) with full spatial context, including in regions of squamous metaplasia. Unsupervised clustering, neighborhood analysis, and manual quantification along the proximal-distal axis characterized the colonic immune landscape, quantified cell-cell interactions, and revealed sex differences. The distal colon was the most affected region during colitis, which was pronounced in males, who exhibited a sex-dependent increase of B cells and reduction of M2-like macrophages. Regions of squamous metaplasia exhibited strong infiltration of numerous immune cell populations, especially in males. Females exhibited more helper T cells and neutrophils at homeostasis and increased M2-like macrophage infiltration in the mid-colon upon colitis. Sex differences were corroborated by plasma cytokine profiles. Our results provide a foundation for future studies of inflammatory intestinal conditions.
Collapse
Affiliation(s)
- Matilda Holm
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna, Sweden
- Division of Biosciences and Nutrition, Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden
| | - Lina Stepanauskaitė
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna, Sweden
- Division of Biosciences and Nutrition, Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden
| | - Anna Bäckström
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna, Sweden
| | - Madeleine Birgersson
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna, Sweden
- Division of Biosciences and Nutrition, Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden
| | - Fabio Socciarelli
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Amena Archer
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna, Sweden
- Division of Biosciences and Nutrition, Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden
| | - Charlotte Stadler
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna, Sweden
| | - Cecilia Williams
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna, Sweden.
- Division of Biosciences and Nutrition, Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden.
| |
Collapse
|
50
|
Yan W, Xiong Y, Lv R, Du C, Yu T, Zhang S, Sui W, Deng S, Xiao J, Xu Y, Zou D, Qiu L, An G. Uncommon biphasic CAR-T expansion induces hemophagocytic lymphohistiocytosis-like syndrome and fatal multiple infections following BCMA CAR-T cell therapy: a case report. J Immunother Cancer 2024; 12:e010080. [PMID: 39608976 PMCID: PMC11603806 DOI: 10.1136/jitc-2024-010080] [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: 07/12/2024] [Accepted: 11/08/2024] [Indexed: 11/30/2024] Open
Abstract
B-cell maturation antigen(BCMA)-directed chimeric antigen receptor (CAR)-T-cell therapy has significantly improved the treatment of relapsed or refractory multiple myeloma (MM). Nevertheless, the uncommon phenomenon of biphasic CAR-T cell expansion in vivo and its related severe toxicities have not been methodically described and studied. Herein, we report a case of patients with MM who experienced two CAR-T cell expansion peaks and subsequently developed multiple severe toxicities following BCMA CAR-T cell infusion. The first expansion peak occurred on Day 7, accompanied by grade 3 cytokine release syndrome. The second peak occurred on Day 28, associated with severe immune effector cell-associated hematotoxicity (ICAHT), immune effector-cell associated hemophagocytic lymphohistiocytosis-like syndrome (IEC-HS), and polymicrobial infections. Both ICAHT and IEC-HS were refractory to our standard treatments; however, human umbilical cord mesenchymal stem cell infusion exhibited some efficacy in improving cytopenia. Despite the administration of a broad-spectrum anti-infective regimen, cytomegalovirus viremia remained uncontrollable, resulting in the development of central nervous system infection, neurological symptoms, and ultimately death. Additionally, we also employed high-dimensional 33-color spectral flow cytometry to describe the dynamic changes in immune cell components and functions between the two expansion peaks. Collectively, this case provides novel insights into the biphasic CAR-T expansion and related immune effector cell-associated toxicities.
Collapse
Affiliation(s)
- Wenqiang Yan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yu Xiong
- Department of Radiation and Medical Oncology for Esophageal Mediastinal and Lymphatic tumors, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Rui Lv
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Chenxing Du
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Tengteng Yu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Shuaishuai Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Weiwei Sui
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Shuhui Deng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Jigang Xiao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yan Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Dehui Zou
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Lugui Qiu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
- Beijing GoBroad Boren Hospital, Beijing, China
| | - Gang An
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
- Beijing GoBroad Boren Hospital, Beijing, China
| |
Collapse
|