1
|
Bosteels V, Janssens S. Striking a balance: new perspectives on homeostatic dendritic cell maturation. Nat Rev Immunol 2024:10.1038/s41577-024-01079-5. [PMID: 39289483 DOI: 10.1038/s41577-024-01079-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2024] [Indexed: 09/19/2024]
Abstract
Dendritic cells (DCs) are crucial gatekeepers of the balance between immunity and tolerance. They exist in two functional states, immature or mature, that refer to an information-sensing versus an information-transmitting state, respectively. Historically, the term DC maturation was used to describe the acquisition of immunostimulatory capacity by DCs following their triggering by pathogens or tissue damage signals. As such, immature DCs were proposed to mediate tolerance, whereas mature DCs were associated with the induction of protective T cell immunity. Later studies have challenged this view and unequivocally demonstrated that two distinct modes of DC maturation exist, homeostatic and immunogenic DC maturation, each with a distinct functional outcome. Therefore, the mere expression of maturation markers cannot be used to predict immunogenicity. How DCs become activated in homeostatic conditions and maintain tolerance remains an area of intense debate. Several recent studies have shed light on the signals driving the homeostatic maturation programme, especially in the conventional type 1 DC (cDC1) compartment. Here, we highlight our growing understanding of homeostatic DC maturation and the relevance of this process for immune tolerance.
Collapse
Affiliation(s)
- Victor Bosteels
- Laboratory for ER Stress and Inflammation, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Sophie Janssens
- Laboratory for ER Stress and Inflammation, VIB Center for Inflammation Research, Ghent, Belgium.
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium.
| |
Collapse
|
2
|
Xia L, Komissarova A, Jacover A, Shovman Y, Arcila-Barrera S, Tornovsky-Babeay S, Jaya Prakashan MM, Nasereddin A, Plaschkes I, Nevo Y, Shiff I, Yosefov-Levi O, Izhiman T, Medvedev E, Eilon E, Wilensky A, Yona S, Parnas O. Systematic identification of gene combinations to target in innate immune cells to enhance T cell activation. Nat Commun 2023; 14:6295. [PMID: 37813864 PMCID: PMC10562403 DOI: 10.1038/s41467-023-41792-8] [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: 07/06/2023] [Accepted: 09/19/2023] [Indexed: 10/11/2023] Open
Abstract
Genetic engineering of immune cells has opened new avenues for improving their functionality but it remains a challenge to pinpoint which genes or combination of genes are the most beneficial to target. Here, we conduct High Multiplicity of Perturbations and Cellular Indexing of Transcriptomes and Epitopes (HMPCITE-seq) to find combinations of genes whose joint targeting improves antigen-presenting cell activity and enhances their ability to activate T cells. Specifically, we perform two genome-wide CRISPR screens in bone marrow dendritic cells and identify negative regulators of CD86, that participate in the co-stimulation programs, including Chd4, Stat5b, Egr2, Med12, and positive regulators of PD-L1, that participate in the co-inhibitory programs, including Sptlc2, Nckap1l, and Pi4kb. To identify the genetic interactions between top-ranked genes and find superior combinations to target, we perform high-order Perturb-Seq experiments and we show that targeting both Cebpb and Med12 results in a better phenotype compared to the single perturbations or other combinations of perturbations.
Collapse
Affiliation(s)
- Lei Xia
- The Lautenberg Center for Immunology and Cancer Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 91120, Israel
| | - Anastasia Komissarova
- The Lautenberg Center for Immunology and Cancer Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 91120, Israel
| | - Arielle Jacover
- The Lautenberg Center for Immunology and Cancer Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 91120, Israel
| | - Yehuda Shovman
- The Lautenberg Center for Immunology and Cancer Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 91120, Israel.
| | - Sebastian Arcila-Barrera
- The Lautenberg Center for Immunology and Cancer Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 91120, Israel
| | - Sharona Tornovsky-Babeay
- The Lautenberg Center for Immunology and Cancer Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 91120, Israel
| | - Milsee Mol Jaya Prakashan
- The Lautenberg Center for Immunology and Cancer Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 91120, Israel
| | - Abdelmajeed Nasereddin
- Core Research Facility, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 91120, Israel
| | - Inbar Plaschkes
- I-CORE Bioinformatics Unit of the Hebrew University and Hadassah Medical Center, Jerusalem, 91120, Israel
| | - Yuval Nevo
- I-CORE Bioinformatics Unit of the Hebrew University and Hadassah Medical Center, Jerusalem, 91120, Israel
| | - Idit Shiff
- Core Research Facility, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 91120, Israel
| | - Oshri Yosefov-Levi
- The Lautenberg Center for Immunology and Cancer Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 91120, Israel
| | - Tamara Izhiman
- The Lautenberg Center for Immunology and Cancer Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 91120, Israel
| | - Eleonora Medvedev
- Core Research Facility, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 91120, Israel
| | - Elad Eilon
- The Lautenberg Center for Immunology and Cancer Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 91120, Israel
| | - Asaf Wilensky
- Department of Periodontology, Hadassah Medical Center, Faculty of Dental Medicine, Hebrew University of Jerusalem, Jerusalem, 91120, Israel
| | - Simon Yona
- The Institute of Biomedical and Oral Research, Hebrew University, Jerusalem, 91120, Israel
| | - Oren Parnas
- The Lautenberg Center for Immunology and Cancer Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 91120, Israel.
| |
Collapse
|
3
|
Zhou R, Liu Y, Wang Z, Lv J, Liao W, Shen Z, Rong X. Nanoparticle-Based MRI-Guided Tumor Microenvironment Heating via the Synergistic Effect of Ferroptosis and Inhibition of TGF-β Signaling. Adv Healthc Mater 2023; 12:e2300176. [PMID: 37093559 DOI: 10.1002/adhm.202300176] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 04/16/2023] [Indexed: 04/25/2023]
Abstract
Although induction of ferroptosis and inhibition of transforming growth factor-β (TGF-β) signaling are both effective ways to reform the tumor microenvironment (TME) and render low-immunogenic tumors responsive to immune checkpoint inhibitor therapy, dose-limiting side effects remain major obstacles hindering their clinical application. Herein, novel sorafenib and anti-TGF-β antibody loaded Fe3 O4 /Gd2 O3 hybrid nanoparticles with conjugation of arginine-glycine-aspartic dimer (FeGd-HN@Sorafenib@TGF-β-antibody@RGD2, FG-STR) are developed. Sorafenib significantly enhances FeGd-HN-triggered ferroptosis and improves maturation and phagocytosis of dendritic cells (DCs) by inducing damage-associated molecular patterns released from ferroptotic cancer cells, while the anti-TGF-β antibody further synergizes with enhanced ferroptosis to promote DC maturation and the recruitment of CD8+ T cells, thus heating the TME. Moreover, the incorporation of RGD2 facilitates the uptake of the FG-STR in tumor cells which lead to a significant dosage reduction of both sorafenib and anti-TGF-β antibody to avoid dose-limiting toxicities. Finally, in vitro and in vivo experiments show that FG-STR has significantly superior intrinsic magnetic resonance imaging (MRI) capability than that of Gadovist, effectively inhibits tumor growth and lung metastasis, and increases the efficacy of anti-programmed cell death-1 treatment. Taken together, this study provides a promising strategy for new advanced MRI-guided TME heating therapies.
Collapse
Affiliation(s)
- Rui Zhou
- Nanfang Hospital, Southern Medical University, Department of Oncology, Guangzhou, Guangdong, 510515, P. R. China
| | - Yantan Liu
- Nanfang Hospital, Southern Medical University, Department of Oncology, Guangzhou, Guangdong, 510515, P. R. China
| | - Zhaojun Wang
- Nanfang Hospital, Southern Medical University, Department of Oncology, Guangzhou, Guangdong, 510515, P. R. China
| | - Jinyu Lv
- Nanfang Hospital, Southern Medical University, Department of Oncology, Guangzhou, Guangdong, 510515, P. R. China
| | - Wangjun Liao
- Nanfang Hospital, Southern Medical University, Department of Oncology, Guangzhou, Guangdong, 510515, P. R. China
| | - Zheyu Shen
- School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Xiaoxiang Rong
- Nanfang Hospital, Southern Medical University, Department of Oncology, Guangzhou, Guangdong, 510515, P. R. China
| |
Collapse
|
4
|
Núñez R, Rodríguez MJ, Lebrón-Martín C, Martín-Astorga MDC, Palomares F, Ramos-Soriano J, Rojo J, Torres MJ, Cañas JA, Mayorga C. Methylation changes induced by a glycodendropeptide immunotherapy and associated to tolerance in mice. Front Immunol 2022; 13:1094172. [PMID: 36643916 PMCID: PMC9832389 DOI: 10.3389/fimmu.2022.1094172] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 11/29/2022] [Indexed: 12/15/2022] Open
Abstract
Introduction Allergen-specific immunotherapy (AIT) is applied as treatment to rise tolerance in patients with food allergies. Although AIT is thoroughly used, the underlying epigenetic events related to tolerant induction are still unknown. Thus, we aim to investigate epigenetic changes that could be related to tolerance in dendritic cells (DCs) from anaphylactic mice to lipid transfer proteins, Pru p 3, in the context of a sublingual immunotherapy (SLIT) with a glycodendropeptide (D1ManPrup3) that has demonstrated tolerant or desensitization responses depending on the treatment dose. Methods Changes in DNA methylation in CpG context were determined comparing Sensitized (Antigen-only) animals and two groups receiving SLIT with the D1ManPrup3 nanostructure (D1ManPrup3-SLIT): Tolerant (2nM D1ManPrup3) and Desensitized (5nM D1ManPrup3), against anaphylactic animals. DNA from lymph nodes-DCs were isolated and then, Whole Genome Bisulphite Sequencing was performed to analyze methylation. Results Most differentially methylated regions were found on the area of influence of gene promoters (DMPRs). Compared to the Anaphylactic group, the highest value was found in Desensitized mice (n = 7,713 DMPRs), followed by Tolerant (n = 4,091 DMPRs) and Sensitized (n = 3,931 DMPRs) mice. Moreover, many of these epigenetic changes were found in genes involved in immune and tolerance responses (Il1b, Il12b, Il1a, Ifng, and Tnf) as shown by functional enrichment (DCs regulation, B cell-mediated immunity, and effector mechanisms). Discussion In conclusion, different doses of D1ManPrup3-SLIT induce different DNA methylation changes, which are reflected in the induction of distinct responses, tolerance, or desensitization.
Collapse
Affiliation(s)
- Rafael Núñez
- Laboratory of Allergy, Allergy Research Group, Instituto de Investigación Biomédica de Málaga-Plataforma Andalusian Centre for Nanomedicine and Biotechnology (IBIMA-BIONAND), Málaga, Spain
| | - María J. Rodríguez
- Laboratory of Allergy, Allergy Research Group, Instituto de Investigación Biomédica de Málaga-Plataforma Andalusian Centre for Nanomedicine and Biotechnology (IBIMA-BIONAND), Málaga, Spain
| | - Clara Lebrón-Martín
- Laboratory of Allergy, Allergy Research Group, Instituto de Investigación Biomédica de Málaga-Plataforma Andalusian Centre for Nanomedicine and Biotechnology (IBIMA-BIONAND), Málaga, Spain
| | - María del Carmen Martín-Astorga
- Laboratory of Allergy, Allergy Research Group, Instituto de Investigación Biomédica de Málaga-Plataforma Andalusian Centre for Nanomedicine and Biotechnology (IBIMA-BIONAND), Málaga, Spain,Department of Medicine, Universidad de Málaga (UMA), Málaga, Spain
| | - Francisca Palomares
- Laboratory of Allergy, Allergy Research Group, Instituto de Investigación Biomédica de Málaga-Plataforma Andalusian Centre for Nanomedicine and Biotechnology (IBIMA-BIONAND), Málaga, Spain
| | - Javier Ramos-Soriano
- Laboratory of Glycosystems, Institute of Chemical Research (IIQ), Spanish National Research Council (CSIC)- Universidad de Sevilla, Sevilla, Spain
| | - Javier Rojo
- Laboratory of Glycosystems, Institute of Chemical Research (IIQ), Spanish National Research Council (CSIC)- Universidad de Sevilla, Sevilla, Spain
| | - María J. Torres
- Laboratory of Allergy, Allergy Research Group, Instituto de Investigación Biomédica de Málaga-Plataforma Andalusian Centre for Nanomedicine and Biotechnology (IBIMA-BIONAND), Málaga, Spain,Department of Medicine, Universidad de Málaga (UMA), Málaga, Spain,Clinical Unit of Allergy, Hospital Regional Universitario de Málaga, Málaga, Spain
| | - José Antonio Cañas
- Laboratory of Allergy, Allergy Research Group, Instituto de Investigación Biomédica de Málaga-Plataforma Andalusian Centre for Nanomedicine and Biotechnology (IBIMA-BIONAND), Málaga, Spain
| | - Cristobalina Mayorga
- Laboratory of Allergy, Allergy Research Group, Instituto de Investigación Biomédica de Málaga-Plataforma Andalusian Centre for Nanomedicine and Biotechnology (IBIMA-BIONAND), Málaga, Spain,Clinical Unit of Allergy, Hospital Regional Universitario de Málaga, Málaga, Spain,*Correspondence: Cristobalina Mayorga,
| |
Collapse
|
5
|
Moshkovits I, Kaminitz A, Reuveni D, Pasmanik‐Chor M, Brazowski E, Mildner A, Leutz A, Zigmond E. Immune cell C/EBPβ deficiency is associated with hepatic mononuclear defects and spontaneous hepatitis but not steatohepatitis induced liver fibrosis. Immun Inflamm Dis 2022; 10:e728. [PMID: 36301029 PMCID: PMC9609438 DOI: 10.1002/iid3.728] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/20/2022] [Accepted: 10/12/2022] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND CCAAT/enhancer-binding protein β (C/EBPβ) is a transcription factor known to be involved in macrophage differentiation and function, steatohepatitis and liver fibrosis. METHODS Immune restricted C/EBPβ deficient and control mice were investigated in steady-state and in the CDA-HFD steatohepatitis model. Mice were assessed for weight change, liver biochemical profile, histology and hepatic phagocytes composition. RESULTS Flow cytometry analysis of hepatic nonparenchymal cells revealed reduced numbers of hepatic monocytes and Kupffer cells and an increase in hepatic MHC class II positive myeloid cells in immune cells restricted C/EBPβ deficient mice. Immune-restricted C/EBPβ deficiency resulted in decreased weight gain and appearance of mild spontaneous liver inflammation. Nevertheless, In the CDA-HFD steatohepatitis model, immune restricted C/EBPβ deficient and proficient mice exhibit similar grade of hepatic steatosis, liver enzymes levels and fibrosis stage. CONCLUSIONS Immune-restricted C/EBPβ deficiency leads to significant alteration in hepatic mononuclear phagocytes composition associated with spontaneous mild hepatitis. Steatohepatitis associated fibrosis is not dependent on C/EBPβ expression by immune cells.
Collapse
Affiliation(s)
- Itay Moshkovits
- Research Center for Digestive DiseasesTel Aviv Sourasky Medical CenterTel AvivIsrael,Sackler Faculty of MedicineTel Aviv UniversityTel AvivIsrael
| | - Ayelet Kaminitz
- Research Center for Digestive DiseasesTel Aviv Sourasky Medical CenterTel AvivIsrael,Sackler Faculty of MedicineTel Aviv UniversityTel AvivIsrael
| | - Debby Reuveni
- Research Center for Digestive DiseasesTel Aviv Sourasky Medical CenterTel AvivIsrael,Sackler Faculty of MedicineTel Aviv UniversityTel AvivIsrael,Center for Autoimmune Liver DiseasesTel Aviv Sourasky Medical CenterTel AvivIsrael
| | - Metsada Pasmanik‐Chor
- Bioinformatics Unit, George‐S. Wise Faculty of Life ScienceTel Aviv UniversityTel AvivIsrael
| | - Eli Brazowski
- Sackler Faculty of MedicineTel Aviv UniversityTel AvivIsrael,Department of PathologyTel Aviv Sourasky Medical CenterTel AvivIsrael
| | | | - Achim Leutz
- Max Delbrück Center for Molecular MedicineBerlinGermany
| | - Ehud Zigmond
- Research Center for Digestive DiseasesTel Aviv Sourasky Medical CenterTel AvivIsrael,Sackler Faculty of MedicineTel Aviv UniversityTel AvivIsrael,Center for Autoimmune Liver DiseasesTel Aviv Sourasky Medical CenterTel AvivIsrael
| |
Collapse
|
6
|
Alsinet C, Primo MN, Lorenzi V, Bello E, Kelava I, Jones CP, Vilarrasa-Blasi R, Sancho-Serra C, Knights AJ, Park JE, Wyspianska BS, Trynka G, Tough DF, Bassett A, Gaffney DJ, Alvarez-Errico D, Vento-Tormo R. Robust temporal map of human in vitro myelopoiesis using single-cell genomics. Nat Commun 2022; 13:2885. [PMID: 35610203 PMCID: PMC9130280 DOI: 10.1038/s41467-022-30557-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 05/06/2022] [Indexed: 11/09/2022] Open
Abstract
Myeloid cells are central to homeostasis and immunity. Characterising in vitro myelopoiesis protocols is imperative for their use in research, immunotherapies, and understanding human myelopoiesis. Here, we generate a >470K cells molecular map of human induced pluripotent stem cells (iPSC) differentiation into macrophages. Integration with in vivo single-cell atlases shows in vitro differentiation recapitulates features of yolk sac hematopoiesis, before definitive hematopoietic stem cells (HSC) emerge. The diversity of myeloid cells generated, including mast cells and monocytes, suggests that HSC-independent hematopoiesis can produce multiple myeloid lineages. We uncover poorly described myeloid progenitors and conservation between in vivo and in vitro regulatory programs. Additionally, we develop a protocol to produce iPSC-derived dendritic cells (DC) resembling cDC2. Using CRISPR/Cas9 knock-outs, we validate the effects of key transcription factors in macrophage and DC ontogeny. This roadmap of myeloid differentiation is an important resource for investigating human fetal hematopoiesis and new therapeutic opportunities.
Collapse
Affiliation(s)
- Clara Alsinet
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK. .,Open Targets, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK.
| | - Maria Nascimento Primo
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK.,Open Targets, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Valentina Lorenzi
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Erica Bello
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK.,Open Targets, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Iva Kelava
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Carla P Jones
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | | | - Carmen Sancho-Serra
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Andrew J Knights
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Jong-Eun Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Korea
| | - Beata S Wyspianska
- Open Targets, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK.,Immunology Research Unit, Medicines Research Centre, GlaxoSmithKline, Stevenage, SG1 2NY, UK
| | - Gosia Trynka
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK.,Open Targets, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - David F Tough
- Immunology Research Unit, Medicines Research Centre, GlaxoSmithKline, Stevenage, SG1 2NY, UK
| | - Andrew Bassett
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK.,Open Targets, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Daniel J Gaffney
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK.
| | - Damiana Alvarez-Errico
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, 08916, Barcelona, Catalonia, Spain.
| | - Roser Vento-Tormo
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK.
| |
Collapse
|
7
|
Gupta YH, Khanom A, Acton SE. Control of Dendritic Cell Function Within the Tumour Microenvironment. Front Immunol 2022; 13:733800. [PMID: 35355992 PMCID: PMC8960065 DOI: 10.3389/fimmu.2022.733800] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 02/09/2022] [Indexed: 12/12/2022] Open
Abstract
The tumour microenvironment (TME) presents a major block to anti-tumour immune responses and to effective cancer immunotherapy. The inflammatory mediators such as cytokines, chemokines, growth factors and prostaglandins generated in the TME alter the phenotype and function of dendritic cells (DCs) that are critical for a successful adaptive immune response against the growing tumour. In this mini review we discuss how tumour cells and the surrounding stroma modulate DC maturation and trafficking to impact T cell function. Fibroblastic stroma and the associated extracellular matrix around tumours can also provide physical restrictions to infiltrating DCs and other leukocytes. We discuss interactions between the inflammatory TME and infiltrating immune cell function, exploring how the inflammatory TME affects generation of T cell-driven anti-tumour immunity. We discuss the open question of the relative importance of antigen-presentation site; locally within the TME versus tumour-draining lymph nodes. Addressing these questions will potentially increase immune surveillance and enhance anti-tumour immunity.
Collapse
Affiliation(s)
- Yukti Hari Gupta
- Stromal Immunology Laboratory, MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | | | - Sophie E. Acton
- Stromal Immunology Laboratory, MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| |
Collapse
|
8
|
Nuñez R, Rodriguez MJ, Palomares F, Gomez F, Jabato FM, Cordoba-Caballero J, Seoane P, Losada J, Rojo J, Torres MJ, Perkins JR, Mayorga C. Transcriptional changes in dendritic cells underlying allergen specific induced tolerance in a mouse model. Sci Rep 2022; 12:2797. [PMID: 35181694 PMCID: PMC8857182 DOI: 10.1038/s41598-022-06186-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 01/14/2022] [Indexed: 12/13/2022] Open
Abstract
To investigate food allergy-tolerance mechanisms induced through allergen-specific immunotherapy we used RNA-Sequencing to measure gene expression in lymph-node-derived dendritic cells from Pru p 3-anaphylactic mice after immunotherapy with glycodendropeptides at 2 nM and 5 nM, leading to permanent tolerance and short-term desensitization, respectively. Gene expression was also measured in mice receiving no immunotherapy (anaphylaxis); and in which anaphylaxis could never occur (antigen-only). Compared to anaphylaxis, the antigen-only group showed the greatest number of expression-changes (411), followed by tolerant (186) and desensitized (119). Only 29 genes changed in all groups, including Il12b, Cebpb and Ifngr1. The desensitized group showed enrichment for genes related to chronic inflammatory response, secretory granule, and regulation of interleukin-12 production; the tolerant group showed genes related to cytokine receptor activity and glucocorticoid receptor binding, suggesting distinct pathways for similar outcomes. We identified genes and processes potentially involved in the restoration of long-term tolerance via allergen-specific immunotherapy, representing potential prognostic biomarkers.
Collapse
Affiliation(s)
- Rafael Nuñez
- Allergy Research Group, Research Laboratory, Allergy Unit, Hospital Regional Universitario de Málaga-IBIMA, Instituto de Investigación Biomédica de Málaga-IBIMA, 29009, Málaga, Spain
| | - Maria Jose Rodriguez
- Allergy Research Group, Research Laboratory, Allergy Unit, Hospital Regional Universitario de Málaga-IBIMA, Instituto de Investigación Biomédica de Málaga-IBIMA, 29009, Málaga, Spain
| | - Francisca Palomares
- Allergy Research Group, Research Laboratory, Allergy Unit, Hospital Regional Universitario de Málaga-IBIMA, Instituto de Investigación Biomédica de Málaga-IBIMA, 29009, Málaga, Spain
| | - Francisca Gomez
- Allergy Clinical Unit, Hospital Regional Universitario de Málaga, Málaga, Spain
| | - Fernando M Jabato
- Department of Molecular Biology and Biochemistry, University of Malaga, Malaga, Spain
| | | | - Pedro Seoane
- Department of Molecular Biology and Biochemistry, University of Malaga, Malaga, Spain
- CIBER de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
| | - Jorge Losada
- Laboratory of Carbohydrates, Instituto de Investigaciones Químicas (IIQ), CSIC-Universidad de Sevilla, Sevilla, Spain
| | - Javier Rojo
- Laboratory of Carbohydrates, Instituto de Investigaciones Químicas (IIQ), CSIC-Universidad de Sevilla, Sevilla, Spain
| | - Maria Jose Torres
- Allergy Research Group, Research Laboratory, Allergy Unit, Hospital Regional Universitario de Málaga-IBIMA, Instituto de Investigación Biomédica de Málaga-IBIMA, 29009, Málaga, Spain
- Allergy Clinical Unit, Hospital Regional Universitario de Málaga, Málaga, Spain
- Nanostructures for Diagnosing and Treatment of Allergic Diseases Laboratory, Centro Andaluz de Nanomedicina y Biotecnología-BIONAND, Málaga, Spain
- Medicine Department, Universidad de Málaga-UMA, Málaga, Spain
| | - James Richard Perkins
- Allergy Research Group, Research Laboratory, Allergy Unit, Hospital Regional Universitario de Málaga-IBIMA, Instituto de Investigación Biomédica de Málaga-IBIMA, 29009, Málaga, Spain
- Department of Molecular Biology and Biochemistry, University of Malaga, Malaga, Spain
- CIBER de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
| | - Cristobalina Mayorga
- Allergy Research Group, Research Laboratory, Allergy Unit, Hospital Regional Universitario de Málaga-IBIMA, Instituto de Investigación Biomédica de Málaga-IBIMA, 29009, Málaga, Spain.
- Allergy Clinical Unit, Hospital Regional Universitario de Málaga, Málaga, Spain.
- Nanostructures for Diagnosing and Treatment of Allergic Diseases Laboratory, Centro Andaluz de Nanomedicina y Biotecnología-BIONAND, Málaga, Spain.
| |
Collapse
|
9
|
Zilio S, Bicciato S, Weed D, Serafini P. CCR1 and CCR5 mediate cancer-induced myelopoiesis and differentiation of myeloid cells in the tumor. J Immunother Cancer 2022; 10:jitc-2021-003131. [PMID: 35064009 PMCID: PMC8785210 DOI: 10.1136/jitc-2021-003131] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/07/2021] [Indexed: 12/21/2022] Open
Abstract
Background Cancer-induced ‘emergency’ myelopoiesis plays a key role in tumor progression by inducing the accumulation of myeloid cells with a suppressive phenotype peripherally and in the tumor. Chemokine receptors (CCRs) and, in particular, CCR1, CCR2, CCR5, and CCR7 are emerging as key regulators of myeloid cell trafficking and function but their precise role has not been completely clarified yet because of the signal redundancy, integration, and promiscuity of chemokines and of the expression of these CCRs on other leukocyte subsets. Methods We used the 4PD nanoparticle for the in vivo targeted silencing of CCR1, CCR2, CCR5, and/or CCR7 in the myeloid cells of tumor bearing mice to evaluate the effect of treatments on tumor growth, myeloid cell trafficking and polarization. We used flow and image cytometry and functional assays to monitor changes in the tumor microenvironment and depletion experiments and immune deficient mice to determine the role of Ly6G+cells during tumor progression. We further evaluated in vitro the impact of chemokine receptor inhibition and tumor derived factors on myeloid cell differentiation from mouse and human hematopoietic stem and precursors cells (HSPCs) using flow cytometry, transcriptome analysis, cytokines beads arrays, functional assays, and mice deficient for CCR1 or CCR5. Results 4PD-mediated in vivo silencing of CCR1 and CCR5 on myeloid cells and myeloid precursors was necessary and sufficient to inhibit tumor progression. Functional studies indicated that this antitumor effect was not mediated by alteration of myeloid cell chemotaxes but rather by the repolarization of polymorphonuclear myeloid-derived suppressor cells (MDSCs) into tumoricidal neutrophils. Transcriptome functional and cytokine analysis indicated that tumor derived factors induced CCL3 and CCL4 in HSPCs that, through the autocrine engagement of CCR1 and CCR5, induced HSPCs differentiation in MDSCs. These finding were confirmed across mice with different genetic backgrounds and using HSPCs from umbilical cord blood and peripheral blood of patients with cancer. Conclusions Our data support the notion that CCR1 and CCR5 and their ligands are a master immunological hub activated by several tumor derived factors. Activation of this pathway is necessary for the differentiation of MDSCs and protumoral macrophages.
Collapse
Affiliation(s)
- Serena Zilio
- Department of Microbiology and Immunology, Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Silvio Bicciato
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Donald Weed
- Department of Otolaryngology, Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Paolo Serafini
- Department of Microbiology and Immunology, Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
10
|
Menzel L, Zschummel M, Crowley T, Franke V, Grau M, Ulbricht C, Hauser A, Siffrin V, Bajénoff M, Acton SE, Akalin A, Lenz G, Willimsky G, Höpken UE, Rehm A. Lymphocyte access to lymphoma is impaired by high endothelial venule regression. Cell Rep 2021; 37:109878. [PMID: 34706240 PMCID: PMC8567313 DOI: 10.1016/j.celrep.2021.109878] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 08/27/2021] [Accepted: 10/01/2021] [Indexed: 12/15/2022] Open
Abstract
Blood endothelial cells display remarkable plasticity depending on the demands of a malignant microenvironment. While studies in solid tumors focus on their role in metabolic adaptations, formation of high endothelial venules (HEVs) in lymph nodes extends their role to the organization of immune cell interactions. As a response to lymphoma growth, blood vessel density increases; however, the fate of HEVs remains elusive. Here, we report that lymphoma causes severe HEV regression in mouse models that phenocopies aggressive human B cell lymphomas. HEV dedifferentiation occurrs as a consequence of a disrupted lymph-carrying conduit system. Mechanosensitive fibroblastic reticular cells then deregulate CCL21 migration paths, followed by deterioration of dendritic cell proximity to HEVs. Loss of this crosstalk deprives HEVs of lymphotoxin-β-receptor (LTβR) signaling, which is indispensable for their differentiation and lymphocyte transmigration. Collectively, this study reveals a remodeling cascade of the lymph node microenvironment that is detrimental for immune cell trafficking in lymphoma.
Collapse
Affiliation(s)
- Lutz Menzel
- Translational Tumorimmunology, Max-Delbrück-Center for Molecular Medicine Berlin, Germany, 13125 Berlin, Germany
| | - Maria Zschummel
- Microenvironmental Regulation in Autoimmunity and Cancer, Max-Delbrück-Center for Molecular Medicine Berlin, 13125 Berlin, Germany
| | - Tadhg Crowley
- Neuroimmunology Laboratory, Max-Delbrück-Center for Molecular Medicine Berlin, Germany, 13125 Berlin, Germany
| | - Vedran Franke
- Bioinformatics & Omics Data Science Platform, BIMSB at Max-Delbrück-Center for Molecular Medicine Berlin, 13125 Berlin, Germany
| | - Michael Grau
- Medical Department A for Hematology, Oncology, and Pneumology, University Hospital Münster, 48149 Münster, Germany
| | - Carolin Ulbricht
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, and Immune Dynamics, Deutsches Rheumaforschungszentrum Berlin, 10117 Berlin, Germany
| | - Anja Hauser
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, and Immune Dynamics, Deutsches Rheumaforschungszentrum Berlin, 10117 Berlin, Germany
| | - Volker Siffrin
- Neuroimmunology Laboratory, Max-Delbrück-Center for Molecular Medicine Berlin, Germany, 13125 Berlin, Germany; Neuroimmunology Laboratory, Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin, 13125 Berlin, Germany
| | - Marc Bajénoff
- Aix Marseille University, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, 13288 Marseille, France
| | - Sophie E Acton
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, WC1E 6BT London, UK
| | - Altuna Akalin
- Bioinformatics & Omics Data Science Platform, BIMSB at Max-Delbrück-Center for Molecular Medicine Berlin, 13125 Berlin, Germany
| | - Georg Lenz
- Medical Department A for Hematology, Oncology, and Pneumology, University Hospital Münster, 48149 Münster, Germany
| | - Gerald Willimsky
- Institute of Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13125 Berlin, Germany; German Cancer Research Center, 69120 Heidelberg, Germany; German Cancer Consortium, partner site Berlin, Germany
| | - Uta E Höpken
- Microenvironmental Regulation in Autoimmunity and Cancer, Max-Delbrück-Center for Molecular Medicine Berlin, 13125 Berlin, Germany
| | - Armin Rehm
- Translational Tumorimmunology, Max-Delbrück-Center for Molecular Medicine Berlin, Germany, 13125 Berlin, Germany.
| |
Collapse
|
11
|
Marongiu L, Protti G, Facchini FA, Valache M, Mingozzi F, Ranzani V, Putignano AR, Salviati L, Bevilacqua V, Curti S, Crosti M, Sarnicola ML, D'Angiò M, Bettini LR, Biondi A, Nespoli L, Tamini N, Clementi N, Mancini N, Abrignani S, Spreafico R, Granucci F. Maturation signatures of conventional dendritic cell subtypes in COVID-19 suggest direct viral sensing. Eur J Immunol 2021; 52:109-122. [PMID: 34333764 PMCID: PMC8420462 DOI: 10.1002/eji.202149298] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 06/09/2021] [Accepted: 07/29/2021] [Indexed: 11/11/2022]
Abstract
Growing evidence suggests that conventional dendritic cells (cDCs) undergo aberrant maturation in COVID‐19, which negatively affects T‐cell activation. The presence of effector T cells in patients with mild disease and dysfunctional T cells in severely ill patients suggests that adequate T‐cell responses limit disease severity. Understanding how cDCs cope with SARS‐CoV‐2 can help elucidate how protective immune responses are generated. Here, we report that cDC2 subtypes exhibit similar infection‐induced gene signatures, with the upregulation of IFN‐stimulated genes and IL‐6 signaling pathways. Furthermore, comparison of cDCs between patients with severe and mild disease showed severely ill patients to exhibit profound downregulation of genes encoding molecules involved in antigen presentation, such as MHCII, TAP, and costimulatory proteins, whereas we observed the opposite for proinflammatory molecules, such as complement and coagulation factors. Thus, as disease severity increases, cDC2s exhibit enhanced inflammatory properties and lose antigen presentation capacity. Moreover, DC3s showed upregulation of anti‐apoptotic genes and accumulated during infection. Direct exposure of cDC2s to the virus in vitro recapitulated the activation profile observed in vivo. Our findings suggest that SARS‐CoV‐2 interacts directly with cDC2s and implements an efficient immune escape mechanism that correlates with disease severity by downregulating crucial molecules required for T‐cell activation.
Collapse
Affiliation(s)
- Laura Marongiu
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy.,National Institute of Molecular Genetics "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Giulia Protti
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy.,National Institute of Molecular Genetics "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Fabio A Facchini
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Mihai Valache
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Francesca Mingozzi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Valeria Ranzani
- National Institute of Molecular Genetics "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Anna Rita Putignano
- National Institute of Molecular Genetics "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Lorenzo Salviati
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy.,National Institute of Molecular Genetics "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Valeria Bevilacqua
- National Institute of Molecular Genetics "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Serena Curti
- National Institute of Molecular Genetics "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Mariacristina Crosti
- National Institute of Molecular Genetics "Romeo ed Enrica Invernizzi", Milan, Italy
| | | | - Mariella D'Angiò
- Pediatric Department and Centro Tettamanti-European Reference Network PaedCan, EuroBloodNet, MetabERN-University of Milano-Bicocca-Fondazione MBBM-Ospedale, San Gerardo, Italy
| | - Laura Rachele Bettini
- Pediatric Department and Centro Tettamanti-European Reference Network PaedCan, EuroBloodNet, MetabERN-University of Milano-Bicocca-Fondazione MBBM-Ospedale, San Gerardo, Italy
| | - Andrea Biondi
- Pediatric Department and Centro Tettamanti-European Reference Network PaedCan, EuroBloodNet, MetabERN-University of Milano-Bicocca-Fondazione MBBM-Ospedale, San Gerardo, Italy
| | - Luca Nespoli
- ASST san Gerardo Hospital, School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Nicolò Tamini
- ASST san Gerardo Hospital, School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Nicola Clementi
- Laboratory of Medical Microbiology and Virology, Vita-Salute San Raffaele University, Milan, Italy.,IRCCS San Raffaele Hospital, Milan, Italy
| | - Nicasio Mancini
- Laboratory of Medical Microbiology and Virology, Vita-Salute San Raffaele University, Milan, Italy.,IRCCS San Raffaele Hospital, Milan, Italy
| | - Sergio Abrignani
- National Institute of Molecular Genetics "Romeo ed Enrica Invernizzi", Milan, Italy.,Department of Clinical Sciences and Community Health, University of Milano, Milan, Italy
| | - Roberto Spreafico
- Institute for Quantitative and Computational Biosciences, University of California, Los Angeles
| | - Francesca Granucci
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy.,National Institute of Molecular Genetics "Romeo ed Enrica Invernizzi", Milan, Italy
| |
Collapse
|
12
|
Menzel L, Höpken UE, Rehm A. Angiogenesis in Lymph Nodes Is a Critical Regulator of Immune Response and Lymphoma Growth. Front Immunol 2020; 11:591741. [PMID: 33343570 PMCID: PMC7744479 DOI: 10.3389/fimmu.2020.591741] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 10/19/2020] [Indexed: 02/06/2023] Open
Abstract
Tumor-induced remodeling of the microenvironment in lymph nodes (LNs) includes the formation of blood vessels, which goes beyond the regulation of metabolism, and shaping a survival niche for tumor cells. In contrast to solid tumors, which primarily rely on neo-angiogenesis, hematopoietic malignancies usually grow within pre-vascularized autochthonous niches in secondary lymphatic organs or the bone marrow. The mechanisms of vascular remodeling in expanding LNs during infection-induced responses have been studied in more detail; in contrast, insights into the conditions of lymphoma growth and lodging remain enigmatic. Based on previous murine studies and clinical trials in human, we conclude that there is not a universal LN-specific angiogenic program applicable. Instead, signaling pathways that are tightly connected to autochthonous and infiltrating cell types contribute variably to LN vascular expansion. Inflammation related angiogenesis within LNs relies on dendritic cell derived pro-inflammatory cytokines stimulating vascular endothelial growth factor-A (VEGF-A) expression in fibroblastic reticular cells, which in turn triggers vessel growth. In high-grade B cell lymphoma, angiogenesis correlates with poor prognosis. Lymphoma cells immigrate and grow in LNs and provide pro-angiogenic growth factors themselves. In contrast to infectious stimuli that impact on LN vasculature, they do not trigger the typical inflammatory and hypoxia-related stroma-remodeling cascade. Blood vessels in LNs are unique in selective recruitment of lymphocytes via high endothelial venules (HEVs). The dissemination routes of neoplastic lymphocytes are usually disease stage dependent. Early seeding via the blood stream requires the expression of the homeostatic chemokine receptor CCR7 and of L-selectin, both cooperate to facilitate transmigration of tumor and also of protective tumor-reactive lymphocytes via HEV structures. In this view, the HEV route is not only relevant for lymphoma cell homing, but also for a continuous immunosurveillance. We envision that HEV functional and structural alterations during lymphomagenesis are not only key to vascular remodeling, but also impact on tumor cell accessibility when targeted by T cell-mediated immunotherapies.
Collapse
Affiliation(s)
- Lutz Menzel
- Translational Tumor Immunology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Uta E. Höpken
- Microenvironmental Regulation in Autoimmunity and Cancer, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Armin Rehm
- Translational Tumor Immunology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| |
Collapse
|