1
|
Warner van Dijk FA, Tong O, O’Neil TR, Bertram KM, Hu K, Baharlou H, Vine EE, Jenns K, Gosselink MP, Toh JW, Papadopoulos T, Barnouti L, Jenkins GJ, Sandercoe G, Haniffa M, Sandgren KJ, Harman AN, Cunningham AL, Nasr N. Characterising plasmacytoid and myeloid AXL+ SIGLEC-6+ dendritic cell functions and their interactions with HIV. PLoS Pathog 2024; 20:e1012351. [PMID: 38924030 PMCID: PMC11233022 DOI: 10.1371/journal.ppat.1012351] [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: 02/15/2024] [Revised: 07/09/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
AXL+ Siglec-6+ dendritic cells (ASDC) are novel myeloid DCs which can be subdivided into CD11c+ and CD123+ expressing subsets. We showed for the first time that these two ASDC subsets are present in inflamed human anogenital tissues where HIV transmission occurs. Their presence in inflamed tissues was supported by single cell RNA analysis of public databases of such tissues including psoriasis diseased skin and colorectal cancer. Almost all previous studies have examined ASDCs as a combined population. Our data revealed that the two ASDC subsets differ markedly in their functions when compared with each other and to pDCs. Relative to their cell functions, both subsets of blood ASDCs but not pDCs expressed co-stimulatory and maturation markers which were more prevalent on CD11c+ ASDCs, thus inducing more T cell proliferation and activation than their CD123+ counterparts. There was also a significant polarisation of naïve T cells by both ASDC subsets toward Th2, Th9, Th22, Th17 and Treg but less toward a Th1 phenotype. Furthermore, we investigated the expression of chemokine receptors that facilitate ASDCs and pDCs migration from blood to inflamed tissues, their HIV binding receptors, and their interactions with HIV and CD4 T cells. For HIV infection, within 2 hours of HIV exposure, CD11c+ ASDCs showed a trend in more viral transfer to T cells than CD123+ ASDCs and pDCs for first phase transfer. However, for second phase transfer, CD123+ ASDCs showed a trend in transferring more HIV than CD11c+ ASDCs and there was no viral transfer from pDCs. As anogenital inflammation is a prerequisite for HIV transmission, strategies to inhibit ASDC recruitment into inflamed tissues and their ability to transmit HIV to CD4 T cells should be considered.
Collapse
Affiliation(s)
- Freja A. Warner van Dijk
- The Westmead Institute for Medical Research, Centre for Virus Research, Westmead, Australia
- The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health, Sydney, Australia
| | - Orion Tong
- The Westmead Institute for Medical Research, Centre for Virus Research, Westmead, Australia
| | - Thomas R. O’Neil
- The Westmead Institute for Medical Research, Centre for Virus Research, Westmead, Australia
- The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health, Sydney, Australia
| | - Kirstie M. Bertram
- The Westmead Institute for Medical Research, Centre for Virus Research, Westmead, Australia
| | - Kevin Hu
- The Westmead Institute for Medical Research, Centre for Virus Research, Westmead, Australia
- The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health, Sydney, Australia
| | - Heeva Baharlou
- The Westmead Institute for Medical Research, Centre for Virus Research, Westmead, Australia
- The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health, Sydney, Australia
| | - Erica E. Vine
- The Westmead Institute for Medical Research, Centre for Virus Research, Westmead, Australia
- The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health, Sydney, Australia
| | - Kate Jenns
- The Westmead Institute for Medical Research, Centre for Virus Research, Westmead, Australia
- The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health, Sydney, Australia
| | | | - James W. Toh
- The Westmead Institute for Medical Research, Centre for Virus Research, Westmead, Australia
- The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health, Sydney, Australia
- Department of Colorectal Surgery, Westmead Hospital, Westmead, Australia
| | - Tim Papadopoulos
- Department of Plastic, Reconstructive, and Aesthetic Surgery, Westmead Private Hospital, Westmead, Australia
| | - Laith Barnouti
- Department of Plastic, Reconstructive, and Aesthetic Surgery, Westmead Private Hospital, Westmead, Australia
| | - Gregory J. Jenkins
- Department of Obstetrics and Gynaecology, Westmead Hospital, Westmead, Australia
| | - Gavin Sandercoe
- Department of Plastic Surgery, Norwest Private Hospital, Bella Vista, Australia
| | - Muzlifah Haniffa
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Knigdom
- Biosciences Institute, Newcastle University, Newcastle, United Knigdom
- Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Knigdom
| | - Kerrie J. Sandgren
- The Westmead Institute for Medical Research, Centre for Virus Research, Westmead, Australia
- The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health, Sydney, Australia
| | - Andrew N. Harman
- The Westmead Institute for Medical Research, Centre for Virus Research, Westmead, Australia
- The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health, Sydney, Australia
| | - Anthony L. Cunningham
- The Westmead Institute for Medical Research, Centre for Virus Research, Westmead, Australia
- The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health, Sydney, Australia
| | - Najla Nasr
- The Westmead Institute for Medical Research, Centre for Virus Research, Westmead, Australia
- The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health, Sydney, Australia
| |
Collapse
|
2
|
Vine EE, Austin PJ, O'Neil TR, Nasr N, Bertram KM, Cunningham AL, Harman AN. Epithelial dendritic cells vs. Langerhans cells: Implications for mucosal vaccines. Cell Rep 2024; 43:113977. [PMID: 38512869 DOI: 10.1016/j.celrep.2024.113977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 02/21/2024] [Accepted: 03/04/2024] [Indexed: 03/23/2024] Open
Abstract
Next-generation vaccines may be delivered via the skin and mucosa. The stratified squamous epithelium (SSE) represents the outermost layer of the skin (epidermis) and type II mucosa (epithelium). Langerhans cells (LCs) have been considered the sole antigen-presenting cells (APCs) to inhabit the SSE; however, it is now clear that dendritic cells (DCs) are also present. Importantly, there are functional differences in how LCs and DCs take up and process pathogens as well as their ability to activate and polarize T cells, though whether DCs participate in neuroimmune interactions like LCs is yet to be elucidated. A correct definition and functional characterization of APCs in the skin and anogenital tissues are of utmost importance for the design of better vaccines and blocking pathogen transmission. Here, we provide a historical perspective on the evolution of our understanding of the APCs that inhabit the SSE, including a detailed review of the most recent literature.
Collapse
Affiliation(s)
- Erica Elizabeth Vine
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW 2145, Australia; Westmead Clinic School, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW 2145, Australia
| | - Paul Jonathon Austin
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW 2145, Australia; School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Westmead, NSW 2145, Australia; Brain and Mind Centre, University of Sydney, Camperdown, NSW 2050, Australia
| | - Thomas Ray O'Neil
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW 2145, Australia; School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Westmead, NSW 2145, Australia
| | - Najla Nasr
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW 2145, Australia; School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Westmead, NSW 2145, Australia
| | - Kirstie Melissa Bertram
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW 2145, Australia; School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Westmead, NSW 2145, Australia
| | - Anthony Lawrence Cunningham
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW 2145, Australia; School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Westmead, NSW 2145, Australia
| | - Andrew Nicholas Harman
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW 2145, Australia; School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Westmead, NSW 2145, Australia.
| |
Collapse
|
3
|
Heger L, Dudziak D, Amon L, Hatscher L, Kaszubowski T, Lehmann CHK. Guidelines for DC preparation and flow cytometric analysis of human lymphohematopoietic tissues. Eur J Immunol 2023; 53:e2249917. [PMID: 36563130 DOI: 10.1002/eji.202249917] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 09/16/2022] [Accepted: 09/19/2022] [Indexed: 12/24/2022]
Abstract
This article is part of the Dendritic Cell Guidelines article series, which provides a collection of state-of-the-art protocols for the preparation, phenotype analysis by flow cytometry, generation, fluorescence microscopy, and functional characterization of mouse and human dendritic cells (DC) from lymphoid organs and various non-lymphoid tissues. Within this article, detailed protocols are presented that allow for the generation of single cell suspensions from human lymphohematopoietic tissues including blood, spleen, thymus, and tonsils with a focus on the subsequent analysis of DC via flow cytometry, as well as flow cytometric cell sorting of primary human DC. Further, prepared single cell suspensions as well as cell sorter-purified DC can be subjected to other applications including cellular enrichment procedures, RNA sequencing, functional assays, and many more. While all protocols were written by experienced scientists who routinely use them in their work, this article was also peer-reviewed by leading experts and approved by all co-authors, making it an essential resource for basic and clinical DC immunologists.
Collapse
Affiliation(s)
- Lukas Heger
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Hartmannstraße 14, D-91052, Erlangen, Germany
| | - Diana Dudziak
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Hartmannstraße 14, D-91052, Erlangen, Germany
- Medical Immunology Campus Erlangen (MICE), D-91054, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), D-91054, Erlangen, Germany
| | - Lukas Amon
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Hartmannstraße 14, D-91052, Erlangen, Germany
| | - Lukas Hatscher
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Hartmannstraße 14, D-91052, Erlangen, Germany
| | - Tomasz Kaszubowski
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Hartmannstraße 14, D-91052, Erlangen, Germany
| | - Christian H K Lehmann
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Hartmannstraße 14, D-91052, Erlangen, Germany
- Medical Immunology Campus Erlangen (MICE), D-91054, Erlangen, Germany
| |
Collapse
|
4
|
O'Neil TR, Harman AN, Cunningham AL, Nasr N, Bertram KM. OMIP-096: A 24-color flow cytometry panel to identify and characterize CD4+ and CD8+ tissue-resident T cells in human skin, intestinal, and type II mucosal tissue. Cytometry A 2023; 103:851-856. [PMID: 37772977 PMCID: PMC10953338 DOI: 10.1002/cyto.a.24782] [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: 09/03/2022] [Revised: 04/06/2023] [Accepted: 07/24/2023] [Indexed: 09/30/2023]
Abstract
There is a great need to understand human immune cells within tissue, where disease manifests and infection occurs. Tissue-resident memory T cells (TRMs) were discovered over a decade ago, there is a great need to understand their role in human disease. We developed a 24-color flow cytometry panel to comprehensively interrogate CD4+ and CD8+ TRMs isolated from human tissues. When interrogating cells within human tissue, enzymatic methods used to liberate cells from within the tissue can cause cleavage of cell surface markers needed to phenotype these cells. Here we carefully select antibody clones and evaluate the effect of enzymatic digestion on the expression of markers relevant to the identification of T cell residency, as well as markers relevant to the activation and immunoregulation status of these cells. We have designed this panel to be applicable across a range of human tissues including skin, intestine, and type II mucosae such as the vagina.
Collapse
Affiliation(s)
- Thomas R. O'Neil
- Centre for Virus Research, The Westmead Institute for Medical ResearchWestmeadAustralia
- The Westmead Clinical School, Faculty of Medicine and HealthThe University of SydneySydneyAustralia
| | - Andrew N. Harman
- Centre for Virus Research, The Westmead Institute for Medical ResearchWestmeadAustralia
- The Westmead Clinical School, Faculty of Medicine and HealthThe University of SydneySydneyAustralia
| | - Anthony L. Cunningham
- Centre for Virus Research, The Westmead Institute for Medical ResearchWestmeadAustralia
- The Westmead Clinical School, Faculty of Medicine and HealthThe University of SydneySydneyAustralia
| | - Najla Nasr
- Centre for Virus Research, The Westmead Institute for Medical ResearchWestmeadAustralia
- The Westmead Clinical School, Faculty of Medicine and HealthThe University of SydneySydneyAustralia
| | - Kirstie M. Bertram
- Centre for Virus Research, The Westmead Institute for Medical ResearchWestmeadAustralia
- The Westmead Clinical School, Faculty of Medicine and HealthThe University of SydneySydneyAustralia
| |
Collapse
|
5
|
Attiogbe E, Larochelle S, Chaib Y, Mainzer C, Mauroux A, Bordes S, Closs B, Gilbert C, Moulin VJ. An in vitro autologous, vascularized, and immunocompetent Tissue Engineered Skin model obtained by the self-assembled approach. Acta Biomater 2023; 168:361-371. [PMID: 37419164 DOI: 10.1016/j.actbio.2023.06.045] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 06/22/2023] [Accepted: 06/28/2023] [Indexed: 07/09/2023]
Abstract
A complete in vitro skin model, containing resident cell types is needed to understand physiology and to consider the role of immune and endothelial cells in dermal drug testing. In this study, a cell extraction technique was developed to isolate resident skin cells from the same human donor while preserving the immune and endothelial cells. Then those cells were used to reconstruct an autologous, vascularized, and immunocompetent Tissue-Engineered Skin model, aviTES. Phenotypic characterization of the viable cells was performed on freshly isolated cells and after thawing through flow cytometry. Dermal cell extracts were characterized as fibroblasts, endothelial and immune cells, and the average amount of each cell type represents 4, 0.5, and 1 million viable cells per g of the dermis, respectively. The 3D models, TES and aviTES, were characterized by a fully differentiated epidermis that showed an increase in the presence of Ki67+ cells in the basolateral layer of the aviTES model. Capillary-like network formation, through the self-assembly of endothelial cells, and the presence of functional immune cells were identified through immunofluorescence staining in aviTES. In addition, the aviTES model was immunocompetent, as evidenced by its capacity to increase the production of pro-inflammatory cytokines TNF-α, MIP-1α, and GM-CSF following LPS stimulation. This study describes an autologous skin model containing a functional resident skin immune system and a capillary network. It provides a relevant tool to study the contribution of the immune system to skin diseases and inflammatory responses and to investigate resident skin cell interactions and drug development. STATEMENT OF SIGNIFICANCE: There is an urgent need for a complete in vitro skin model containing the resident cell types to better understand the role of immune and endothelial cells in skin and to be able to use it for drug testing. Actual 3D models of human skin most often contain only fibroblasts and keratinocytes with a limited number of models containing endothelial cells or a limited variety of immune cells. This study describes an autologous skin model containing a functional resident skin immune system and a capillary network. It provides a relevant tool to study the contribution of the immune system to skin diseases and inflammatory responses and to investigate interactions between resident skin cell, improving our capacity to develop new drugs.
Collapse
Affiliation(s)
- Emilie Attiogbe
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval (LOEX), Québec, QC, Canada; Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Sébastien Larochelle
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval (LOEX), Québec, QC, Canada; Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Yanis Chaib
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval (LOEX), Québec, QC, Canada; Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | | | | | | | | | - Caroline Gilbert
- Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Véronique J Moulin
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval (LOEX), Québec, QC, Canada; Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Faculty of Medicine, Université Laval, Québec, QC, Canada.
| |
Collapse
|
6
|
Morin S, Bélanger S, Cortez Ghio S, Pouliot R. Eicosapentaenoic acid reduces the proportion of IL-17A-producing T cells in a 3D psoriatic skin model. J Lipid Res 2023; 64:100428. [PMID: 37597582 PMCID: PMC10509711 DOI: 10.1016/j.jlr.2023.100428] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 08/03/2023] [Accepted: 08/11/2023] [Indexed: 08/21/2023] Open
Abstract
Psoriasis is a skin disease presenting as erythematous lesions with accentuated proliferation of epidermal keratinocytes, infiltration of leukocytes, and dysregulated lipid metabolism. T cells play essential roles in the disease. n-3 polyunsaturated fatty acids are anti-inflammatory metabolites, which exert an immunosuppressive effect on healthy T cells. However, the precise mechanistic processes of n-3 polyunsaturated fatty acids on T cells in psoriasis are still unrevealed. In this study, we aimed to evaluate the action of eicosapentaenoic acid (EPA) on T cells in a psoriatic skin model produced with T cells. A coculture of psoriatic keratinocytes and polarized T cells was prepared using culture media, which was either supplemented with 10 μM EPA or left unsupplemented. Healthy and psoriatic skin substitutes were produced according to the self-assembly method. In the coculture model, EPA reduced the proportion of IL-17A-positive cells, while increasing that of FOXP3-positive cells, suggesting an increase in the polarization of regulatory T cells. In the 3D psoriatic skin model, EPA normalized the proliferation of psoriatic keratinocytes and diminished the levels of IL-17A. The expression of the proteins of the signal transducer and activator of transcription was influenced following EPA supplementation with downregulation of the phosphorylation levels of signal transducer and activator of transcription 3 in the dermis. Finally, the NFκB signaling pathway was modified in the EPA-supplemented substitutes with an increase in Fas amounts. Ultimately, our results suggest that in this psoriatic model, EPA exerts its anti-inflammatory action by decreasing the proportion of IL-17A-producing T cells.
Collapse
Affiliation(s)
- Sophie Morin
- Center for Research in Experimental Organogenesis of Laval University/LOEX, Regenerative Medicine Axis, CHU of Quebec/Laval University Research Center, Qu ebec, QC, Canada; Faculty of Pharmacy, Laval University, Quebec, QC, Canada
| | - Sarah Bélanger
- Center for Research in Experimental Organogenesis of Laval University/LOEX, Regenerative Medicine Axis, CHU of Quebec/Laval University Research Center, Qu ebec, QC, Canada; Faculty of Pharmacy, Laval University, Quebec, QC, Canada
| | | | - Roxane Pouliot
- Center for Research in Experimental Organogenesis of Laval University/LOEX, Regenerative Medicine Axis, CHU of Quebec/Laval University Research Center, Qu ebec, QC, Canada; Faculty of Pharmacy, Laval University, Quebec, QC, Canada.
| |
Collapse
|
7
|
Lischetti U, Tastanova A, Singer F, Grob L, Carrara M, Cheng PF, Martínez Gómez JM, Sella F, Haunerdinger V, Beisel C, Levesque MP. Dynamic thresholding and tissue dissociation optimization for CITE-seq identifies differential surface protein abundance in metastatic melanoma. Commun Biol 2023; 6:830. [PMID: 37563418 PMCID: PMC10415364 DOI: 10.1038/s42003-023-05182-6] [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: 07/25/2022] [Accepted: 07/26/2023] [Indexed: 08/12/2023] Open
Abstract
Multi-omics profiling by CITE-seq bridges the RNA-protein gap in single-cell analysis but has been largely applied to liquid biopsies. Applying CITE-seq to clinically relevant solid biopsies to characterize healthy tissue and the tumor microenvironment is an essential next step in single-cell translational studies. In this study, gating of cell populations based on their transcriptome signatures for use in cell type-specific ridge plots allowed identification of positive antibody signals and setting of manual thresholds. Next, we compare five skin dissociation protocols by taking into account dissociation efficiency, captured cell type heterogeneity and recovered surface proteome. To assess the effect of enzymatic digestion on transcriptome and epitope expression in immune cell populations, we analyze peripheral blood mononuclear cells (PBMCs) with and without dissociation. To further assess the RNA-protein gap, RNA-protein we perform codetection and correlation analyses on thresholded protein values. Finally, in a proof-of-concept study, using protein abundance analysis on selected surface markers in a cohort of healthy skin, primary, and metastatic melanoma we identify CD56 surface marker expression on metastatic melanoma cells, which was further confirmed by multiplex immunohistochemistry. This work provides practical guidelines for processing and analysis of clinically relevant solid tissue biopsies for biomarker discovery.
Collapse
Affiliation(s)
- Ulrike Lischetti
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058, Basel, Switzerland
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031, Basel, Switzerland
| | - Aizhan Tastanova
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| | - Franziska Singer
- ETH Zurich, NEXUS Personalized Health Technologies, Wagistrasse 18, 8952, Schlieren, Switzerland
- SIB Swiss Institute of Bioinformatics, Zurich, Switzerland
| | - Linda Grob
- ETH Zurich, NEXUS Personalized Health Technologies, Wagistrasse 18, 8952, Schlieren, Switzerland
- SIB Swiss Institute of Bioinformatics, Zurich, Switzerland
| | - Matteo Carrara
- ETH Zurich, NEXUS Personalized Health Technologies, Wagistrasse 18, 8952, Schlieren, Switzerland
- SIB Swiss Institute of Bioinformatics, Zurich, Switzerland
| | - Phil F Cheng
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Julia M Martínez Gómez
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Federica Sella
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Veronika Haunerdinger
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Christian Beisel
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058, Basel, Switzerland
| | - Mitchell P Levesque
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
8
|
Bertram KM, O'Neil TR, Vine EE, Baharlou H, Cunningham AL, Harman AN. Defining the landscape of human epidermal mononuclear phagocytes. Immunity 2023; 56:459-460. [PMID: 36921567 DOI: 10.1016/j.immuni.2023.02.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 11/08/2022] [Accepted: 02/03/2023] [Indexed: 03/16/2023]
Affiliation(s)
- Kirstie M Bertram
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia; The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health, Westmead, NSW, Australia; The University of Sydney, Sydney Infectious Diseases, Faculty of Medicine and Health, Westmead, NSW, Australia; Centre for Immunology and Allergy Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia
| | - Thomas R O'Neil
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia; The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health, Westmead, NSW, Australia; The University of Sydney, Sydney Infectious Diseases, Faculty of Medicine and Health, Westmead, NSW, Australia
| | - Erica E Vine
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia; The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health, Westmead, NSW, Australia; The University of Sydney, Sydney Infectious Diseases, Faculty of Medicine and Health, Westmead, NSW, Australia
| | - Heeva Baharlou
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia; The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health, Westmead, NSW, Australia; The University of Sydney, Sydney Infectious Diseases, Faculty of Medicine and Health, Westmead, NSW, Australia
| | - Anthony L Cunningham
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia; The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health, Westmead, NSW, Australia; The University of Sydney, Sydney Infectious Diseases, Faculty of Medicine and Health, Westmead, NSW, Australia
| | - Andrew N Harman
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia; The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health, Westmead, NSW, Australia; The University of Sydney, Sydney Infectious Diseases, Faculty of Medicine and Health, Westmead, NSW, Australia; Centre for Immunology and Allergy Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.
| |
Collapse
|
9
|
Murray-Brown W, Guo Y, Small A, Lowe K, Weedon H, Smith MD, Lester SE, Proudman SM, Rao NL, Hao LY, Nagpal S, Wechalekar MD. Differential expansion of T peripheral helper cells in early rheumatoid arthritis and osteoarthritis synovium. RMD Open 2022; 8:rmdopen-2022-002563. [PMID: 36270740 PMCID: PMC9594577 DOI: 10.1136/rmdopen-2022-002563] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 10/03/2022] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVES Programmed cell death protein 1 (PD-1)-expressing T cells are implicated in the pathogenesis of autoimmune inflammatory diseases such as rheumatoid arthritis. A subset of CXCR5- T cells, termed T peripheral helper (Tph) cells, which drive B cell differentiation, have been identified in ectopic lymphoid structures in established rheumatoid arthritis synovial tissue. Here, we aimed to characterise these in treatment-naïve, early rheumatoid arthritis to determine whether these cells accumulate prior to fully established disease. METHODS Fresh dissociated tissue and peripheral blood mononuclear cell (PBMC) suspensions were stained with Zombie UV, followed by anti-CD45RO, PD-1, CD3, ICOS, CD8, CD4, CD20, CXCR5, TIGIT and CD38 antibodies prior to analysis. For histology, rheumatoid arthritis synovial sections were prepared for Opal multispectral immunofluorescence with anti-CD45RO, CD20, PD-1 and CXCR5 antibodies. Images were acquired on the Perkin Elmer Vectra V.3.0 imaging system and analysed using InForm Advanced Image Analysis software. RESULTS Flow cytometry revealed T cell infiltration in the rheumatoid arthritis synovium with differential expression of PD-1, CD45RO, ICOS, TIGIT and CD38. We observed a higher frequency of PD1hiCXCR5- Tph in rheumatoid arthritis synovial tissue and PBMCs versus controls, and no significant difference in T follicular helper cell frequency. Microscopy identified a 10-fold increase of Tph cells in early rheumatoid arthritis synovial follicular and diffuse regions, and identified Tph adjacent to germinal centre B cells. CONCLUSIONS These data demonstrate that PD-1hi Tph cells are present in early rheumatoid arthritis, but not osteoarthritis synovium, and therefore may provide a target for treatment of patients with early rheumatoid arthritis.
Collapse
Affiliation(s)
- William Murray-Brown
- Department of Rheumatology, Flinders University, Bedford Park, South Australia, Australia
| | - Yanxia Guo
- Discovery Immunology, Janssen Research and Development, Spring House, Pennsylvania, USA
| | - Annabelle Small
- Department of Rheumatology, Flinders University, Bedford Park, South Australia, Australia
| | - Katie Lowe
- Department of Rheumatology, Flinders University, Bedford Park, South Australia, Australia
| | - Helen Weedon
- Department of Rheumatology, Flinders University, Bedford Park, South Australia, Australia
| | - Malcolm D Smith
- Department of Rheumatology, Flinders University, Bedford Park, South Australia, Australia
| | - Susan E Lester
- Rheumatology Research Group, Basil Hetzel Institute for Medical Research, Woodville South, South Australia, Australia
| | - Susanna M Proudman
- Department of Rheumatology, Royal Adelaide Hospital, Adelaide, South Australia, Australia,Discipline of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
| | - Navin L Rao
- Discovery Immunology, Janssen Research and Development, Spring House, Pennsylvania, USA
| | - Ling-Yang Hao
- Discovery Immunology, Janssen Research and Development, Spring House, Pennsylvania, USA
| | - Sunil Nagpal
- Discovery Immunology, Janssen Research and Development, Spring House, Pennsylvania, USA
| | - Mihir D Wechalekar
- Department of Rheumatology, Flinders University, Bedford Park, South Australia, Australia,Department of Rheumatology, Flinders Medical Centre, Bedford Park, South Australia, Australia
| |
Collapse
|
10
|
Burja B, Paul D, Tastanova A, Edalat SG, Gerber R, Houtman M, Elhai M, Bürki K, Staeger R, Restivo G, Lang R, Sodin-Semrl S, Lakota K, Tomšič M, Levesque MP, Distler O, Rotar Ž, Robinson MD, Frank-Bertoncelj M. An Optimized Tissue Dissociation Protocol for Single-Cell RNA Sequencing Analysis of Fresh and Cultured Human Skin Biopsies. Front Cell Dev Biol 2022; 10:872688. [PMID: 35573685 PMCID: PMC9096112 DOI: 10.3389/fcell.2022.872688] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 04/13/2022] [Indexed: 12/16/2022] Open
Abstract
We present an optimized dissociation protocol for preparing high-quality skin cell suspensions for in-depth single-cell RNA-sequencing (scRNA-seq) analysis of fresh and cultured human skin. Our protocol enabled the isolation of a consistently high number of highly viable skin cells from small freshly dissociated punch skin biopsies, which we use for scRNA-seq studies. We recapitulated not only the main cell populations of existing single-cell skin atlases, but also identified rare cell populations, such as mast cells. Furthermore, we effectively isolated highly viable single cells from ex vivo cultured skin biopsy fragments and generated a global single-cell map of the explanted human skin. The quality metrics of the generated scRNA-seq datasets were comparable between freshly dissociated and cultured skin. Overall, by enabling efficient cell isolation and comprehensive cell mapping, our skin dissociation-scRNA-seq workflow can greatly facilitate scRNA-seq discoveries across diverse human skin pathologies and ex vivo skin explant experimentations.
Collapse
Affiliation(s)
- Blaž Burja
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Dominique Paul
- Department of Molecular Life Sciences and Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland
| | - Aizhan Tastanova
- Department of Dermatology, University of Zurich, University Hospital Zurich, Schlieren, Switzerland
| | - Sam G. Edalat
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Reto Gerber
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Molecular Life Sciences and Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland
| | - Miranda Houtman
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Muriel Elhai
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Kristina Bürki
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Ramon Staeger
- Department of Dermatology, University of Zurich, University Hospital Zurich, Schlieren, Switzerland
| | - Gaetana Restivo
- Department of Dermatology, University of Zurich, University Hospital Zurich, Schlieren, Switzerland
| | - Ramon Lang
- Department of Dermatology, University of Zurich, University Hospital Zurich, Schlieren, Switzerland
| | - Snezna Sodin-Semrl
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Katja Lakota
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Matija Tomšič
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Mitchell P. Levesque
- Department of Dermatology, University of Zurich, University Hospital Zurich, Schlieren, Switzerland
| | - Oliver Distler
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Žiga Rotar
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Mark D. Robinson
- Department of Molecular Life Sciences and Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland
| | - Mojca Frank-Bertoncelj
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- BioMed X Institute, Heidelberg, Germany
| |
Collapse
|
11
|
Marsh SE, Walker AJ, Kamath T, Dissing-Olesen L, Hammond TR, de Soysa TY, Young AMH, Murphy S, Abdulraouf A, Nadaf N, Dufort C, Walker AC, Lucca LE, Kozareva V, Vanderburg C, Hong S, Bulstrode H, Hutchinson PJ, Gaffney DJ, Hafler DA, Franklin RJM, Macosko EZ, Stevens B. Dissection of artifactual and confounding glial signatures by single-cell sequencing of mouse and human brain. Nat Neurosci 2022; 25:306-316. [PMID: 35260865 PMCID: PMC11645269 DOI: 10.1038/s41593-022-01022-8] [Citation(s) in RCA: 210] [Impact Index Per Article: 70.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 01/25/2022] [Indexed: 12/31/2022]
Abstract
A key aspect of nearly all single-cell sequencing experiments is dissociation of intact tissues into single-cell suspensions. While many protocols have been optimized for optimal cell yield, they have often overlooked the effects that dissociation can have on ex vivo gene expression. Here, we demonstrate that use of enzymatic dissociation on brain tissue induces an aberrant ex vivo gene expression signature, most prominently in microglia, which is prevalent in published literature and can substantially confound downstream analyses. To address this issue, we present a rigorously validated protocol that preserves both in vivo transcriptional profiles and cell-type diversity and yield across tissue types and species. We also identify a similar signature in postmortem human brain single-nucleus RNA-sequencing datasets, and show that this signature is induced in freshly isolated human tissue by exposure to elevated temperatures ex vivo. Together, our results provide a methodological solution for preventing artifactual gene expression changes during fresh tissue digestion and a reference for future deeper analysis of the potential confounding states present in postmortem human samples.
Collapse
Affiliation(s)
- Samuel E Marsh
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Alec J Walker
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Tushar Kamath
- Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Lasse Dissing-Olesen
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Timothy R Hammond
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - T Yvanka de Soysa
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Adam M H Young
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Sarah Murphy
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
| | - Abdulraouf Abdulraouf
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Naeem Nadaf
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Connor Dufort
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
| | - Alicia C Walker
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
| | - Liliana E Lucca
- Department of Neurology and Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Velina Kozareva
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Charles Vanderburg
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Soyon Hong
- UK Dementia Research Institute, University College London, London, UK
| | - Harry Bulstrode
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Peter J Hutchinson
- Department of Clinical Neurosciences, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | | | - David A Hafler
- Department of Neurology and Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Robin J M Franklin
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Evan Z Macosko
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Beth Stevens
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
12
|
Sato T, Ogawa Y, Ishikawa A, Nagasaka Y, Kinoshita M, Shiokawa I, Shimada S, Momosawa A, Kawamura T. Revisiting the Experimental Methods for Human Skin T Cell Analysis. JID INNOVATIONS 2022; 2:100125. [PMID: 35620704 PMCID: PMC9127406 DOI: 10.1016/j.xjidi.2022.100125] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 02/05/2022] [Accepted: 03/04/2022] [Indexed: 10/27/2022] Open
|
13
|
Han L, Jara CP, Wang O, Shi Y, Wu X, Thibivilliers S, Wóycicki RK, Carlson MA, Velander WH, Araújo EP, Libault M, Zhang C, Lei Y. Isolating and cryopreserving pig skin cells for single-cell RNA sequencing study. PLoS One 2022; 17:e0263869. [PMID: 35176067 PMCID: PMC8853494 DOI: 10.1371/journal.pone.0263869] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 01/29/2022] [Indexed: 11/27/2022] Open
Abstract
The pig skin architecture and physiology are similar to those of humans. Thus, the pig model is very valuable for studying skin biology and testing therapeutics. The single-cell RNA sequencing (scRNA-seq) technology allows quantitatively analyzing cell types, compositions, states, signaling, and receptor-ligand interactome at single-cell resolution and at high throughput. scRNA-seq has been used to study mouse and human skins. However, studying pig skin with scRNA-seq is still rare. A critical step for successful scRNA-seq is to obtain high-quality single cells from the pig skin tissue. Here we report a robust method for isolating and cryopreserving pig skin single cells for scRNA-seq. We showed that pig skin could be efficiently dissociated into single cells with high cell viability using the Miltenyi Human Whole Skin Dissociation kit and the Miltenyi gentleMACS Dissociator. Furthermore, the obtained single cells could be cryopreserved using 90% FBS + 10% DMSO without causing additional cell death, cell aggregation, or changes in gene expression profiles. Using the developed protocol, we were able to identify all the major skin cell types. The protocol and results from this study are valuable for the skin research scientific community.
Collapse
Affiliation(s)
- Li Han
- School of Biological Science, University of Nebraska, Lincoln, Nebraska, United States of America
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, Nebraska, United States of America
- Department of Biomedical Engineering, Pennsylvania State University, University Park, Pennsylvania, United States of America
- Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Carlos P. Jara
- Nursing School, University of Campinas, Campinas SP, Brazil
- Laboratory of Cell Signaling, University of Campinas, Campinas SP, Brazil
| | - Ou Wang
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, Nebraska, United States of America
| | - Yu Shi
- School of Biological Science, University of Nebraska, Lincoln, Nebraska, United States of America
| | - Xinran Wu
- Department of Biomedical Engineering, Pennsylvania State University, University Park, Pennsylvania, United States of America
- Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Sandra Thibivilliers
- Department of Agronomy and Horticulture, Center for Plant Science Innovation, University of Nebraska, Lincoln, Nebraska, United States of America
| | - Rafał K. Wóycicki
- Department of Agronomy and Horticulture, Center for Plant Science Innovation, University of Nebraska, Lincoln, Nebraska, United States of America
| | - Mark A. Carlson
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Department of Surgery, University of Nebraska Medical Center and the VA Nebraska-Western Iowa Health Care System, Omaha, Nebraska, United States of America
| | - William H. Velander
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, Nebraska, United States of America
| | - Eliana P. Araújo
- Nursing School, University of Campinas, Campinas SP, Brazil
- Laboratory of Cell Signaling, University of Campinas, Campinas SP, Brazil
| | - Marc Libault
- Department of Agronomy and Horticulture, Center for Plant Science Innovation, University of Nebraska, Lincoln, Nebraska, United States of America
| | - Chi Zhang
- School of Biological Science, University of Nebraska, Lincoln, Nebraska, United States of America
| | - Yuguo Lei
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, Nebraska, United States of America
- Department of Biomedical Engineering, Pennsylvania State University, University Park, Pennsylvania, United States of America
- Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, United States of America
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Department of Surgery, University of Nebraska Medical Center and the VA Nebraska-Western Iowa Health Care System, Omaha, Nebraska, United States of America
- Sartorius Mammalian Cell Culture Facility, Pennsylvania State University, University Park, Pennsylvania, United States of America
| |
Collapse
|
14
|
Doyle CM, Fewings NL, Ctercteko G, Byrne SN, Harman AN, Bertram KM. OMIP 082: A 25-color phenotyping to define human innate lymphoid cells, natural killer cells, mucosal-associated invariant T cells, and γδ T cells from freshly isolated human intestinal tissue. Cytometry A 2022; 101:196-202. [PMID: 35018731 DOI: 10.1002/cyto.a.24529] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/18/2021] [Accepted: 12/22/2021] [Indexed: 11/12/2022]
Abstract
We developed a 25-color flow cytometry panel to comprehensively interrogate innate lymphoid cells (ILC), mucosal-associated invariant T (MAIT) cells, natural killer (NK) cells and γδ T cells in human tissues. The ability to isolate and interrogate these cells from fresh human tissue is crucial in understanding the role these cells play at immune-privileged mucosal surfaces like the intestine in health and disease settings. However, liberating these cells from tissue is extremely challenging as many key surface identification markers are susceptible to enzymatic cleavage. Choosing the correct enzyme-antibody clone combination within a high-parameter panel is, therefore, a critical consideration. Here, we present a comprehensive, in-depth analysis of the effect different common digestive enzyme blends have on key surface markers used to identify these cell types. In addition, we compared multiple antibody clones for surface markers that are highly susceptible to enzymatic cleavage, such as CD127 and NKp44, to achieve the most consistent and superior staining patterns among donors.
Collapse
Affiliation(s)
- Chloe M Doyle
- Centre for Immunology and Allergy Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia.,Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia.,Faculty of Medicine and Health, The University of Sydney, School of Medical Sciences, Westmead, New South Wales, Australia.,Faculty of Medicine and Health, The University of Sydney, Westmead Clinical School, Westmead, New South Wales, Australia
| | - Nicole L Fewings
- Centre for Immunology and Allergy Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia.,Faculty of Medicine and Health, The University of Sydney, School of Medical Sciences, Westmead, New South Wales, Australia.,Faculty of Medicine and Health, The University of Sydney, Westmead Clinical School, Westmead, New South Wales, Australia
| | - Grahame Ctercteko
- Centre for Immunology and Allergy Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia.,Faculty of Medicine and Health, The University of Sydney, Westmead Clinical School, Westmead, New South Wales, Australia.,Department of Colorectal Surgery, Westmead Hospital, Westmead, New South Wales, Australia
| | - Scott N Byrne
- Centre for Immunology and Allergy Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia.,Faculty of Medicine and Health, The University of Sydney, School of Medical Sciences, Westmead, New South Wales, Australia
| | - Andrew N Harman
- Centre for Immunology and Allergy Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia.,Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia.,Faculty of Medicine and Health, The University of Sydney, School of Medical Sciences, Westmead, New South Wales, Australia
| | - Kirstie M Bertram
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia.,Faculty of Medicine and Health, The University of Sydney, School of Medical Sciences, Westmead, New South Wales, Australia.,Faculty of Medicine and Health, The University of Sydney, Westmead Clinical School, Westmead, New South Wales, Australia
| |
Collapse
|
15
|
HIV transmitting mononuclear phagocytes; integrating the old and new. Mucosal Immunol 2022; 15:542-550. [PMID: 35173293 PMCID: PMC9259493 DOI: 10.1038/s41385-022-00492-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/24/2022] [Accepted: 01/27/2022] [Indexed: 02/04/2023]
Abstract
In tissue, mononuclear phagocytes (MNP) are comprised of Langerhans cells, dendritic cells, macrophages and monocyte-derived cells. They are the first immune cells to encounter HIV during transmission and transmit the virus to CD4 T cells as a consequence of their antigen presenting cell function. To understand the role these cells play in transmission, their phenotypic and functional characterisation is important. With advancements in high parameter single cell technologies, new MNPs subsets are continuously being discovered and their definition and classification is in a state of flux. This has important implications for our knowledge of HIV transmission, which requires a deeper understanding to design effective vaccines and better blocking strategies. Here we review the historical research of the role MNPs play in HIV transmission up to the present day and revaluate these studies in the context of our most recent understandings of the MNP system.
Collapse
|
16
|
Wu CH, Lin HH, Wu YY, Chiu YL, Huang LY, Cheng CC, Yang CC, Tsai TN. Generation of Functional Cardiomyocytes from Human Gastric Fibroblast-Derived Induced Pluripotent Stem Cells. Biomedicines 2021; 9:biomedicines9111565. [PMID: 34829794 PMCID: PMC8615619 DOI: 10.3390/biomedicines9111565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/23/2021] [Accepted: 10/26/2021] [Indexed: 11/23/2022] Open
Abstract
Coronary artery diseases are major problems of the world. Coronary artery disease patients frequently suffer from peptic ulcers when they receive aspirin treatment. For diagnostic and therapeutic purposes, the implementation of panendoscopy (PES) with biopsy is necessary. Some biopsy samples are wasted after the assay is completed. In the present study, we established a protocol for human gastric fibroblast isolation and induced pluripotent stem cell (iPSC) generation from gastric fibroblasts via PES with biopsy. We showed that these iPSCs can be differentiated into functional cardiomyocytes in vitro. To our knowledge, this is the first study to generate iPSCs from gastric fibroblasts in vitro.
Collapse
Affiliation(s)
- Chih-Hsien Wu
- Department of Biochemistry, National Defense Medical Center, Taipei 11490, Taiwan; (C.-H.W.); (Y.-L.C.)
- Division of Cardiology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan; (L.-Y.H.); (C.-C.C.); (C.-C.Y.)
| | - Hsuan-Hwai Lin
- Division of Gastroenterology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Centre, Taipei 11490, Taiwan;
| | - Yi-Ying Wu
- Division of Hematology/Oncology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan;
| | - Yi-Lin Chiu
- Department of Biochemistry, National Defense Medical Center, Taipei 11490, Taiwan; (C.-H.W.); (Y.-L.C.)
| | - Li-Yen Huang
- Division of Cardiology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan; (L.-Y.H.); (C.-C.C.); (C.-C.Y.)
- Division of Cardiology, Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan County 32551, Taiwan
| | - Cheng-Chung Cheng
- Division of Cardiology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan; (L.-Y.H.); (C.-C.C.); (C.-C.Y.)
| | - Chung-Chi Yang
- Division of Cardiology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan; (L.-Y.H.); (C.-C.C.); (C.-C.Y.)
- Division of Cardiology, Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan County 32551, Taiwan
| | - Tsung-Neng Tsai
- Division of Cardiology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan; (L.-Y.H.); (C.-C.C.); (C.-C.Y.)
- Correspondence:
| |
Collapse
|
17
|
Doyle CM, Vine EE, Bertram KM, Baharlou H, Rhodes JW, Dervish S, Gosselink MP, Di Re A, Collins GP, Reza F, Toh JWT, Pathma-Nathan N, Ahlenstiel G, Ctercteko G, Cunningham AL, Harman AN, Byrne SN. Optimal Isolation Protocols for Examining and Interrogating Mononuclear Phagocytes From Human Intestinal Tissue. Front Immunol 2021; 12:727952. [PMID: 34566985 PMCID: PMC8462295 DOI: 10.3389/fimmu.2021.727952] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 08/11/2021] [Indexed: 12/12/2022] Open
Abstract
The human intestine contains numerous mononuclear phagocytes (MNP), including subsets of conventional dendritic cells (cDC), macrophages (Mf) and monocytes, each playing their own unique role within the intestinal immune system and homeostasis. The ability to isolate and interrogate MNPs from fresh human tissue is crucial if we are to understand the role of these cells in homeostasis, disease settings and immunotherapies. However, liberating these cells from tissue is problematic as many of the key surface identification markers they express are susceptible to enzymatic cleavage and they are highly susceptible to cell death. In addition, the extraction process triggers immunological activation/maturation which alters their functional phenotype. Identifying the evolving, complex and highly heterogenous repertoire of MNPs by flow cytometry therefore requires careful selection of digestive enzyme blends that liberate viable cells and preserve recognition epitopes involving careful selection of antibody clones to enable analysis and sorting for functional assays. Here we describe a method for the anatomical separation of mucosa and submucosa as well as isolating lymphoid follicles from human jejunum, ileum and colon. We also describe in detail the optimised enzyme digestion methods needed to acquire functionally immature and biologically functional intestinal MNPs. A comprehensive list of screened antibody clones is also presented which allows for the development of high parameter flow cytometry panels to discriminate all currently identified human tissue MNP subsets including pDCs, cDC1, cDC2 (langerin+ and langerin-), newly described DC3, monocytes, Mf1, Mf2, Mf3 and Mf4. We also present a novel method to account for autofluorescent signal from tissue macrophages. Finally, we demonstrate that these methods can successfully be used to sort functional, immature intestinal DCs that can be used for functional assays such as cytokine production assays.
Collapse
Affiliation(s)
- Chloe M Doyle
- Centre for Immunology and Allergy Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia.,Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia
| | - Erica E Vine
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia.,Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia
| | - Kirstie M Bertram
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia.,Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia
| | - Heeva Baharlou
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia
| | - Jake W Rhodes
- Centre for Immunology and Allergy Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia
| | - Suat Dervish
- Westmead Cytometry, The Westmead Institute for Medical Research, Westmead, NSW, Australia
| | - Martijn P Gosselink
- Centre for Immunology and Allergy Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia.,Department of Colorectal Surgery, Westmead Hospital, Westmead, NSW, Australia
| | - Angelina Di Re
- Centre for Immunology and Allergy Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia.,Department of Colorectal Surgery, Westmead Hospital, Westmead, NSW, Australia
| | - Geoffrey P Collins
- Centre for Immunology and Allergy Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia.,Department of Colorectal Surgery, Westmead Hospital, Westmead, NSW, Australia
| | - Faizur Reza
- Centre for Immunology and Allergy Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia.,Department of Colorectal Surgery, Westmead Hospital, Westmead, NSW, Australia
| | - James W T Toh
- Centre for Immunology and Allergy Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia.,Department of Colorectal Surgery, Westmead Hospital, Westmead, NSW, Australia
| | - Nimalan Pathma-Nathan
- Centre for Immunology and Allergy Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia.,Department of Colorectal Surgery, Westmead Hospital, Westmead, NSW, Australia
| | - Golo Ahlenstiel
- Storr Liver Centre, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Blacktown Clinical School, Western Sydney University, Blacktown, NSW, Australia.,Blacktown Hospital, Western Sydney Local Area Health District (WSLHD), Blacktown, NSW, Australia
| | - Grahame Ctercteko
- Centre for Immunology and Allergy Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia.,Department of Colorectal Surgery, Westmead Hospital, Westmead, NSW, Australia
| | - Anthony L Cunningham
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia
| | - Andrew N Harman
- Centre for Immunology and Allergy Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia
| | - Scott N Byrne
- Centre for Immunology and Allergy Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia
| |
Collapse
|
18
|
Zirbes A, Joseph J, Lopez JC, Sayaman RW, Basam M, Seewaldt VL, LaBarge MA. Changes in Immune Cell Types with Age in Breast are Consistent with a Decline in Immune Surveillance and Increased Immunosuppression. J Mammary Gland Biol Neoplasia 2021; 26:247-261. [PMID: 34341887 PMCID: PMC8566425 DOI: 10.1007/s10911-021-09495-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/19/2021] [Indexed: 12/22/2022] Open
Abstract
A majority of breast cancers (BC) are age-related and we seek to determine what cellular and molecular changes occur in breast tissue with age that make women more susceptible to cancer initiation. Immune-epithelial cell interactions are important during mammary gland development and the immune system plays an important role in BC progression. The composition of human immune cell populations is known to change in peripheral blood with age and in breast tissue during BC progression. Less is known about changes in immune populations in normal breast tissue and how their interactions with mammary epithelia change with age. We quantified densities of T cells, B cells, and macrophage subsets in pathologically normal breast tissue from 122 different women who ranged in age from 24 to 74 years old. Donor-matched peripheral blood from a subset of 20 donors was analyzed by flow cytometry. Tissue immune cell densities and localizations relative to the epithelium were quantified in situ with machine learning-based image analyses of multiplex immunohistochemistry-stained tissue sections. In situ results were corroborated with flow cytometry analyses of peri-epithelial immune cells from primary breast tissue preparations and transcriptome analyses of public data from bulk tissue reduction mammoplasties. Proportions of immune cell subsets in breast tissue and donor-matched peripheral blood were not correlated. Density (cells/mm2) of T and B lymphocytes in situ decreased with age. T cells and macrophages preferentially localized near or within epithelial bilayers, rather than the intralobular stroma. M2 macrophage density was higher than M1 macrophage density and this difference was due to higher density of M2 in the intralobular stroma. Transcriptional signature analyses suggested age-dependent decline in adaptive immune cell populations and functions and increased innate immune cell activity. T cells and macrophages are so intimately associated with the epithelia that they are embedded within the bilayer, suggesting an important role for immune-epithelial cell interactions. Age-associated decreased T cell density in peri-epithelial regions, and increased M2 macrophage density in intralobular stroma suggests the emergence of a tissue microenvironment that is simultaneously immune-senescent and immunosuppressive with age.
Collapse
Affiliation(s)
- Arrianna Zirbes
- Department of Population Sciences, Beckman Research Institute, City of Hope National Medical Center, 1500 E Duarte Road, Duarte, CA, 91010, USA
- Irell and Manella Graduate School of Biological Sciences, City of Hope, Duarte, CA, USA
| | - Jesuchristopher Joseph
- Department of Population Sciences, Beckman Research Institute, City of Hope National Medical Center, 1500 E Duarte Road, Duarte, CA, 91010, USA
| | - Jennifer C Lopez
- Department of Population Sciences, Beckman Research Institute, City of Hope National Medical Center, 1500 E Duarte Road, Duarte, CA, 91010, USA
| | - Rosalyn W Sayaman
- Department of Population Sciences, Beckman Research Institute, City of Hope National Medical Center, 1500 E Duarte Road, Duarte, CA, 91010, USA
- Center for Cancer and Aging, Beckman Research Institute, City of Hope, Duarte, CA, USA
- Cancer Metabolism Training Program, Beckman Research Institute, City of Hope, Duarte, CA, USA
- Department of Laboratory Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Mudaser Basam
- Department of Population Sciences, Beckman Research Institute, City of Hope National Medical Center, 1500 E Duarte Road, Duarte, CA, 91010, USA
| | - Victoria L Seewaldt
- Department of Population Sciences, Beckman Research Institute, City of Hope National Medical Center, 1500 E Duarte Road, Duarte, CA, 91010, USA
| | - Mark A LaBarge
- Department of Population Sciences, Beckman Research Institute, City of Hope National Medical Center, 1500 E Duarte Road, Duarte, CA, 91010, USA.
- Center for Cancer and Aging, Beckman Research Institute, City of Hope, Duarte, CA, USA.
- Centre for Cancer Biomarkers CCBIO, University of Bergen, Bergen, Norway.
| |
Collapse
|
19
|
Schunkert EM, Shah PN, Divito SJ. Skin Resident Memory T Cells May Play Critical Role in Delayed-Type Drug Hypersensitivity Reactions. Front Immunol 2021; 12:654190. [PMID: 34497600 PMCID: PMC8419326 DOI: 10.3389/fimmu.2021.654190] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 08/06/2021] [Indexed: 12/31/2022] Open
Abstract
Delayed-type drug hypersensitivity reactions (dtDHR) are immune-mediated reactions with skin and visceral manifestations ranging from mild to severe. Clinical care is negatively impacted by a limited understanding of disease pathogenesis. Though T cells are believed to orchestrate disease, the type of T cell and the location and mechanism of T cell activation remain unknown. Resident memory T cells (TRM) are a unique T cell population potentially well situated to act as key mediators in disease pathogenesis, but significant obstacles to defining, identifying, and testing TRM in dtDHR preclude definitive conclusions at this time. Deeper mechanistic interrogation to address these unanswered questions is necessary, as involvement of TRM in disease has significant implications for prediction, diagnosis, and treatment of disease.
Collapse
|
20
|
Marsh‐Wakefield FMD, Mitchell AJ, Norton SE, Ashhurst TM, Leman JKH, Roberts JM, Harte JE, McGuire HM, Kemp RA. Making the most of high-dimensional cytometry data. Immunol Cell Biol 2021; 99:680-696. [PMID: 33797774 PMCID: PMC8453896 DOI: 10.1111/imcb.12456] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 01/03/2023]
Abstract
High-dimensional cytometry represents an exciting new era of immunology research, enabling the discovery of new cells and prediction of patient responses to therapy. A plethora of analysis and visualization tools and programs are now available for both new and experienced users; however, the transition from low- to high-dimensional cytometry requires a change in the way users think about experimental design and data analysis. Data from high-dimensional cytometry experiments are often underutilized, because of both the size of the data and the number of possible combinations of markers, as well as to a lack of understanding of the processes required to generate meaningful data. In this article, we explain the concepts behind designing high-dimensional cytometry experiments and provide considerations for new and experienced users to design and carry out high-dimensional experiments to maximize quality data collection.
Collapse
Affiliation(s)
- Felix MD Marsh‐Wakefield
- Vascular Immunology UnitDiscipline of PathologyThe University of SydneySydneyNSWAustralia
- Charles Perkins CentreThe University of SydneySydneyNSWAustralia
- School of Medical SciencesFaculty of Medicine and HealthThe University of SydneySydneyNSWAustralia
| | - Andrew J Mitchell
- Department of Chemical EngineeringMaterials Characterisation and Fabrication PlatformThe University of MelbourneParkvilleVICAustralia
| | - Samuel E Norton
- Nanix LtdDunedinNew Zealand
- Department of Microbiology and ImmunologyUniversity of OtagoDunedinNew Zealand
| | - Thomas Myles Ashhurst
- Charles Perkins CentreThe University of SydneySydneyNSWAustralia
- Sydney CytometryUniversity of SydneySydneyNSWAustralia
- Ramaciotti Facility for Human Systems BiologyThe University of SydneySydneyNSWAustralia
| | - Julia KH Leman
- Department of Microbiology and ImmunologyUniversity of OtagoDunedinNew Zealand
| | | | - Jessica E Harte
- Department of Microbiology and ImmunologyUniversity of OtagoDunedinNew Zealand
| | - Helen M McGuire
- Charles Perkins CentreThe University of SydneySydneyNSWAustralia
- Ramaciotti Facility for Human Systems BiologyThe University of SydneySydneyNSWAustralia
- Translational Immunology GroupDiscipline of PathologyThe University of SydneySydneyNSWAustralia
| | - Roslyn A Kemp
- Department of Microbiology and ImmunologyUniversity of OtagoDunedinNew Zealand
| |
Collapse
|
21
|
Targeting human langerin promotes HIV-1 specific humoral immune responses. PLoS Pathog 2021; 17:e1009749. [PMID: 34324611 PMCID: PMC8354475 DOI: 10.1371/journal.ppat.1009749] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 08/10/2021] [Accepted: 06/24/2021] [Indexed: 12/01/2022] Open
Abstract
The main avenue for the development of an HIV-1 vaccine remains the induction of protective antibodies. A rationale approach is to target antigen to specific receptors on dendritic cells (DC) via fused monoclonal antibodies (mAb). In mouse and non-human primate models, targeting of skin Langerhans cells (LC) with anti-Langerin mAbs fused with HIV-1 Gag antigen drives antigen-specific humoral responses. The development of these immunization strategies in humans requires a better understanding of early immune events driven by human LC. We therefore produced anti-Langerin mAbs fused with the HIV-1 gp140z Envelope (αLC.Env). First, we show that primary skin human LC and in vitro differentiated LC induce differentiation and expansion of naïve CD4+ T cells into T follicular helper (Tfh) cells. Second, when human LC are pre-treated with αLC.Env, differentiated Tfh cells significantly promote the production of specific IgG by B cells. Strikingly, HIV-Env-specific Ig are secreted by HIV-specific memory B cells. Consistently, we found that receptors and cytokines involved in Tfh differentiation and B cell functions are upregulated by LC during their maturation and after targeting Langerin. Finally, we show that subcutaneous immunization of mice by αLC.Env induces germinal center (GC) reaction in draining lymph nodes with higher numbers of Tfh cells, Env-specific B cells, as well as specific IgG serum levels compared to mice immunized with the non-targeting Env antigen. Altogether, we provide evidence that human LC properly targeted may be licensed to efficiently induce Tfh cell and B cell responses in GC. In recent years, the place of innovative vaccines based on the induction/regulation and modulation of the immune response with the aim to elicit an integrated T- and B cell immune responses against complex antigens has emerged besides “classical” vaccine vectors. Targeting antigens to dendritic cells is a vaccine technology concept supported by more than a decade of animal models and human pre-clinical experimentation. Recent investigations in animals underscored that Langerhans cells (LC) are an important target to consider for the induction of antibody responses by DC targeting vaccine approaches. Nonetheless, the development of these immunization strategies in humans remains elusive. We therefore developed and produced an HIV vaccine candidate targeting specifically LC through the Langerin receptor. We tested the ability of our vaccine candidate of targeting LC from skin explant and of inducing in vitro the differentiation of T follicular helper (Tfh) cells. Using complementary in vitro models, we demonstrated that Tfh cells induced by human LC are functional and the targeting of LC by our vaccine candidate promotes the secretion of anti-HIV IgG by memory B cells from HIV-infected individuals. In this study human LC exhibit key cellular functions able to drive potent anti-HIV-1 humoral responses providing mechanistic evidence of the Tfh- and B cell stimulating functions of primary skin targeted LC. Finally, we demonstrated in Xcr1DTA mice the significant advantage of LC targeting for inducing Tfh and germinal center (GC)-B cells and anti-HIV-1 antibodies. Therefore, the targeting of the human Langerin receptor appears to be a promising strategy for developing efficient HIV-1 vaccine.
Collapse
|
22
|
Bertram KM, Truong NR, Smith JB, Kim M, Sandgren KJ, Feng KL, Herbert JJ, Rana H, Danastas K, Miranda-Saksena M, Rhodes JW, Patrick E, Cohen RC, Lim J, Merten SL, Harman AN, Cunningham AL. Herpes Simplex Virus type 1 infects Langerhans cells and the novel epidermal dendritic cell, Epi-cDC2s, via different entry pathways. PLoS Pathog 2021; 17:e1009536. [PMID: 33905459 PMCID: PMC8104422 DOI: 10.1371/journal.ppat.1009536] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 05/07/2021] [Accepted: 04/06/2021] [Indexed: 12/24/2022] Open
Abstract
Skin mononuclear phagocytes (MNPs) provide the first interactions of invading viruses with the immune system. In addition to Langerhans cells (LCs), we recently described a second epidermal MNP population, Epi-cDC2s, in human anogenital epidermis that is closely related to dermal conventional dendritic cells type 2 (cDC2) and can be preferentially infected by HIV. Here we show that in epidermal explants topically infected with herpes simplex virus (HSV-1), both LCs and Epi-cDC2s interact with HSV-1 particles and infected keratinocytes. Isolated Epi-cDC2s support higher levels of infection than LCs in vitro, inhibited by acyclovir, but both MNP subtypes express similar levels of the HSV entry receptors nectin-1 and HVEM, and show similar levels of initial uptake. Using inhibitors of endosomal acidification, actin and cholesterol, we found that HSV-1 utilises different entry pathways in each cell type. HSV-1 predominantly infects LCs, and monocyte-derived MNPs, via a pH-dependent pathway. In contrast, Epi-cDC2s are mainly infected via a pH-independent pathway which may contribute to the enhanced infection of Epi-cDC2s. Both cells underwent apoptosis suggesting that Epi-cDC2s may follow the same dermal migration and uptake by dermal MNPs that we have previously shown for LCs. Thus, we hypothesize that the uptake of HSV and infection of Epi-cDC2s will stimulate immune responses via a different pathway to LCs, which in future may help guide HSV vaccine development and adjuvant targeting.
Collapse
Affiliation(s)
- Kirstie M. Bertram
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, Australia
- The Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Naomi R. Truong
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, Australia
- The Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Jacinta B. Smith
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, Australia
- The Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Min Kim
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, Australia
- The Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Kerrie J. Sandgren
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, Australia
- The Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Konrad L. Feng
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, Australia
- The Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Jason J. Herbert
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, Australia
- The Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Hafsa Rana
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, Australia
- The Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Kevin Danastas
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, Australia
- The Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Monica Miranda-Saksena
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, Australia
- The Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Jake W. Rhodes
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, Australia
- The Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Ellis Patrick
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, Australia
- The School of Mathematics and Statistics, Faculty of Science, The University of Sydney, Camperdown, Australia
| | - Ralph C. Cohen
- Department of Surgery, University of Sydney and The Children’s Hospital at Westmead, Westmead, Australia
| | - Jake Lim
- Department of Surgery, Westmead Private Hospital, Westmead, Australia
| | - Steven L. Merten
- Department of Surgery, Macquarie University Hospital, Macquarie Park, Australia
| | - Andrew N. Harman
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, Australia
- The School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Westmead, Australia
| | - Anthony L. Cunningham
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, Australia
- The Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
- * E-mail:
| |
Collapse
|
23
|
Rhodes JW, Botting RA, Bertram KM, Vine EE, Rana H, Baharlou H, Vegh P, O'Neil TR, Ashhurst AS, Fletcher J, Parnell GP, Graham JD, Nasr N, Lim JJK, Barnouti L, Haertsch P, Gosselink MP, Di Re A, Reza F, Ctercteko G, Jenkins GJ, Brooks AJ, Patrick E, Byrne SN, Hunter E, Haniffa MA, Cunningham AL, Harman AN. Human anogenital monocyte-derived dendritic cells and langerin+cDC2 are major HIV target cells. Nat Commun 2021; 12:2147. [PMID: 33846309 PMCID: PMC8042121 DOI: 10.1038/s41467-021-22375-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 03/12/2021] [Indexed: 02/06/2023] Open
Abstract
Tissue mononuclear phagocytes (MNP) are specialised in pathogen detection and antigen presentation. As such they deliver HIV to its primary target cells; CD4 T cells. Most MNP HIV transmission studies have focused on epithelial MNPs. However, as mucosal trauma and inflammation are now known to be strongly associated with HIV transmission, here we examine the role of sub-epithelial MNPs which are present in a diverse array of subsets. We show that HIV can penetrate the epithelial surface to interact with sub-epithelial resident MNPs in anogenital explants and define the full array of subsets that are present in the human anogenital and colorectal tissues that HIV may encounter during sexual transmission. In doing so we identify two subsets that preferentially take up HIV, become infected and transmit the virus to CD4 T cells; CD14+CD1c+ monocyte-derived dendritic cells and langerin-expressing conventional dendritic cells 2 (cDC2).
Collapse
Affiliation(s)
- Jake W Rhodes
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Westmead Clinical School, Faculty of Medicine and Health Sydney, The University of Sydney, Westmead, NSW, Australia
| | - Rachel A Botting
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Westmead Clinical School, Faculty of Medicine and Health Sydney, The University of Sydney, Westmead, NSW, Australia.,Biosciences Institute, The University of Newcastle, Newcastle upon Tyne, UK
| | - Kirstie M Bertram
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Westmead Clinical School, Faculty of Medicine and Health Sydney, The University of Sydney, Westmead, NSW, Australia
| | - Erica E Vine
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Westmead Clinical School, Faculty of Medicine and Health Sydney, The University of Sydney, Westmead, NSW, Australia
| | - Hafsa Rana
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Westmead Clinical School, Faculty of Medicine and Health Sydney, The University of Sydney, Westmead, NSW, Australia
| | - Heeva Baharlou
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Westmead Clinical School, Faculty of Medicine and Health Sydney, The University of Sydney, Westmead, NSW, Australia
| | - Peter Vegh
- Biosciences Institute, The University of Newcastle, Newcastle upon Tyne, UK
| | - Thomas R O'Neil
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Westmead Clinical School, Faculty of Medicine and Health Sydney, The University of Sydney, Westmead, NSW, Australia
| | - Anneliese S Ashhurst
- School of Medical Sciences, Faculty of Medicine and Health Sydney, The University of Sydney, Westmead, NSW, Australia
| | - James Fletcher
- Biosciences Institute, The University of Newcastle, Newcastle upon Tyne, UK
| | - Grant P Parnell
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Westmead Clinical School, Faculty of Medicine and Health Sydney, The University of Sydney, Westmead, NSW, Australia
| | - J Dinny Graham
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Westmead Clinical School, Faculty of Medicine and Health Sydney, The University of Sydney, Westmead, NSW, Australia
| | - Najla Nasr
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,School of Medical Sciences, Faculty of Medicine and Health Sydney, The University of Sydney, Westmead, NSW, Australia
| | | | | | - Peter Haertsch
- Burns Unit, Concord Repatriation General Hospital, Sydney, Australia
| | - Martijn P Gosselink
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Department of Colorectal Surgery, Westmead Hospital, Westmead, NSW, Australia
| | - Angelina Di Re
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Department of Colorectal Surgery, Westmead Hospital, Westmead, NSW, Australia
| | - Faizur Reza
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Department of Colorectal Surgery, Westmead Hospital, Westmead, NSW, Australia
| | - Grahame Ctercteko
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Department of Colorectal Surgery, Westmead Hospital, Westmead, NSW, Australia
| | - Gregory J Jenkins
- Department of Obstetrics and Gynaecology, Westmead Hospital, Westmead, NSW, Australia
| | - Andrew J Brooks
- Department of Urology, Westmead Hospital, Westmead, NSW, Australia
| | - Ellis Patrick
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,School of Maths and Statistics, Faculty of Science, The University of Sydney, Camperdown, NSW, Australia
| | - Scott N Byrne
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,School of Medical Sciences, Faculty of Medicine and Health Sydney, The University of Sydney, Westmead, NSW, Australia
| | | | - Muzlifah A Haniffa
- Biosciences Institute, The University of Newcastle, Newcastle upon Tyne, UK.,Wellcome Sanger Institute, Hinxton, UK.,Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Anthony L Cunningham
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,Westmead Clinical School, Faculty of Medicine and Health Sydney, The University of Sydney, Westmead, NSW, Australia
| | - Andrew N Harman
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia. .,School of Medical Sciences, Faculty of Medicine and Health Sydney, The University of Sydney, Westmead, NSW, Australia.
| |
Collapse
|
24
|
Abstract
Dendritic cell (DC) vaccines are a safe and effective means of inducing tumor immune responses, however, a better understanding of DC biology is required in order to realize their full potential. Recent advances in DC biology have identified a crucial role for cDC1 in tumor immune responses, making this DC subset an attractive vaccine target. Human cDC1 exclusively express the C-type-lectin-like receptor, CLEC9A (DNGR-1) that plays an important role in cross-presentation, the process by which effective CD8+ T cell responses are generated. CLEC9A antibodies deliver antigen specifically to cDC1 for the induction of humoral, CD4+ and CD8+ T cell responses and are therefore promising candidates to develop as vaccines for infectious diseases and cancer. The development of human CLEC9A antibodies now facilitates their application as vaccines for cancer immunotherapy. Here we discuss the recent advances in CLEC9A targeting antibodies as vaccines for cancer and their translation to the clinic.
Collapse
Affiliation(s)
- M H Lahoud
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Australia
| | - K J Radford
- Cancer Immunotherapies Laboratory, Mater Research Institute, University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Australia
| |
Collapse
|
25
|
Reynolds G, Vegh P, Fletcher J, Poyner EFM, Stephenson E, Goh I, Botting RA, Huang N, Olabi B, Dubois A, Dixon D, Green K, Maunder D, Engelbert J, Efremova M, Polański K, Jardine L, Jones C, Ness T, Horsfall D, McGrath J, Carey C, Popescu DM, Webb S, Wang XN, Sayer B, Park JE, Negri VA, Belokhvostova D, Lynch MD, McDonald D, Filby A, Hagai T, Meyer KB, Husain A, Coxhead J, Vento-Tormo R, Behjati S, Lisgo S, Villani AC, Bacardit J, Jones PH, O'Toole EA, Ogg GS, Rajan N, Reynolds NJ, Teichmann SA, Watt FM, Haniffa M. Developmental cell programs are co-opted in inflammatory skin disease. Science 2021; 371:eaba6500. [PMID: 33479125 PMCID: PMC7611557 DOI: 10.1126/science.aba6500] [Citation(s) in RCA: 310] [Impact Index Per Article: 77.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 09/03/2020] [Accepted: 12/01/2020] [Indexed: 12/16/2022]
Abstract
The skin confers biophysical and immunological protection through a complex cellular network established early in embryonic development. We profiled the transcriptomes of more than 500,000 single cells from developing human fetal skin, healthy adult skin, and adult skin with atopic dermatitis and psoriasis. We leveraged these datasets to compare cell states across development, homeostasis, and disease. Our analysis revealed an enrichment of innate immune cells in skin during the first trimester and clonal expansion of disease-associated lymphocytes in atopic dermatitis and psoriasis. We uncovered and validated in situ a reemergence of prenatal vascular endothelial cell and macrophage cellular programs in atopic dermatitis and psoriasis lesional skin. These data illustrate the dynamism of cutaneous immunity and provide opportunities for targeting pathological developmental programs in inflammatory skin diseases.
Collapse
Affiliation(s)
- Gary Reynolds
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Peter Vegh
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - James Fletcher
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Elizabeth F M Poyner
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne NE2 4LP, UK
| | - Emily Stephenson
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Issac Goh
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Rachel A Botting
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Ni Huang
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Bayanne Olabi
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Department of Dermatology, NHS Lothian, Lauriston Building, Edinburgh EH3 9EN, UK
| | - Anna Dubois
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne NE2 4LP, UK
| | - David Dixon
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Kile Green
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Daniel Maunder
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Justin Engelbert
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Mirjana Efremova
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Krzysztof Polański
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Laura Jardine
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Claire Jones
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Thomas Ness
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Dave Horsfall
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Jim McGrath
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Christopher Carey
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Dorin-Mirel Popescu
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Simone Webb
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Xiao-Nong Wang
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Ben Sayer
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Jong-Eun Park
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Victor A Negri
- Centre for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital Campus, London SE1 9RT, UK
| | - Daria Belokhvostova
- Centre for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital Campus, London SE1 9RT, UK
| | - Magnus D Lynch
- Centre for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital Campus, London SE1 9RT, UK
| | - David McDonald
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Andrew Filby
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Tzachi Hagai
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Kerstin B Meyer
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Akhtar Husain
- Department of Pathology, Royal Victoria Infirmary, Newcastle upon Tyne NE1 4LP, UK
| | - Jonathan Coxhead
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Roser Vento-Tormo
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Sam Behjati
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
- Department of Paediatrics, University of Cambridge, Cambridge CB2 0SP, UK
| | - Steven Lisgo
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Alexandra-Chloé Villani
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Jaume Bacardit
- School of Computing, Newcastle University, Newcastle upon Tyne NE4 5TG, UK
| | - Philip H Jones
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
- MRC Cancer Unit, University of Cambridge, Cambridge CB2 0XZ, UK
| | - Edel A O'Toole
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Queen Mary University of London, London, UK
| | - Graham S Ogg
- MRC Human Immunology Unit, Oxford Biomedical Research Centre, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Neil Rajan
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne NE2 4LP, UK
| | - Nick J Reynolds
- Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne NE2 4LP, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK.
- Theory of Condensed Matter Group, Cavendish Laboratory/Department of Physics, University of Cambridge, Cambridge CB3 0HE, UK
| | - Fiona M Watt
- Centre for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital Campus, London SE1 9RT, UK.
| | - Muzlifah Haniffa
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK.
- Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne NE2 4LP, UK
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| |
Collapse
|
26
|
Oulee A, Ma F, Teles RMB, de Andrade Silva BJ, Pellegrini M, Klechevsky E, Harman AN, Rhodes JW, Modlin RL. Identification of Genes Encoding Antimicrobial Proteins in Langerhans Cells. Front Immunol 2021; 12:695373. [PMID: 34512625 PMCID: PMC8426439 DOI: 10.3389/fimmu.2021.695373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 08/06/2021] [Indexed: 12/03/2022] Open
Abstract
Langerhans cells (LCs) reside in the epidermis where they are poised to mount an antimicrobial response against microbial pathogens invading from the outside environment. To elucidate potential pathways by which LCs contribute to host defense, we mined published LC transcriptomes deposited in GEO and the scientific literature for genes that participate in antimicrobial responses. Overall, we identified 31 genes in LCs that encode proteins that contribute to antimicrobial activity, ten of which were cross-validated in at least two separate experiments. Seven of these ten antimicrobial genes encode chemokines, CCL1, CCL17, CCL19, CCL2, CCL22, CXCL14 and CXCL2, which mediate both antimicrobial and inflammatory responses. Of these, CCL22 was detected in seven of nine transcriptomes and by PCR in cultured LCs. Overall, the antimicrobial genes identified in LCs encode proteins with broad antibacterial activity, including against Staphylococcus aureus, which is the leading cause of skin infections. Thus, this study illustrates that LCs, consistent with their anatomical location, are programmed to mount an antimicrobial response against invading pathogens in skin.
Collapse
Affiliation(s)
- Aislyn Oulee
- Division of Dermatology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Feiyang Ma
- Division of Dermatology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, United States.,Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Rosane M B Teles
- Division of Dermatology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Bruno J de Andrade Silva
- Division of Dermatology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Eynav Klechevsky
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| | - Andrew N Harman
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,School of Medical Sciences, Faculty of Medicine and Health Sydney, The University of Sydney, Westmead, NSW, Australia
| | - Jake W Rhodes
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia.,School of Medical Sciences, Faculty of Medicine and Health Sydney, The University of Sydney, Westmead, NSW, Australia
| | - Robert L Modlin
- Division of Dermatology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
27
|
Kröger M, Scheffel J, Nikolaev VV, Shirshin EA, Siebenhaar F, Schleusener J, Lademann J, Maurer M, Darvin ME. In vivo non-invasive staining-free visualization of dermal mast cells in healthy, allergy and mastocytosis humans using two-photon fluorescence lifetime imaging. Sci Rep 2020; 10:14930. [PMID: 32913196 PMCID: PMC7484787 DOI: 10.1038/s41598-020-71901-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 08/17/2020] [Indexed: 12/12/2022] Open
Abstract
Mast cells (MCs) are multifunctional cells of the immune system and are found in skin and all major tissues of the body. They contribute to the pathology of several diseases including urticaria, psoriasis, atopic dermatitis and mastocytosis where they are increased at lesional sites. Histomorphometric analysis of skin biopsies serves as a routine method for the assessment of MC numbers and their activation status, which comes with major limitations. As of now, non-invasive techniques to study MCs in vivo are not available. Here, we describe a label-free imaging technique to visualize MCs and their activation status in the human papillary dermis in vivo. This technique uses two-photon excited fluorescence lifetime imaging (TPE-FLIM) signatures, which are different for MCs and other dermal components. TPE-FLIM allows for the visualization and quantification of dermal MCs in healthy subjects and patients with skin diseases. Moreover, TPE-FLIM can differentiate between two MC populations in the papillary dermis in vivo-resting and activated MCs with a sensitivity of 0.81 and 0.87 and a specificity of 0.85 and 0.84, respectively. Results obtained on healthy volunteers and allergy and mastocytosis patients indicate the existence of other MC subpopulations within known resting and activated MC populations. The developed method may become an important tool for non-invasive in vivo diagnostics and therapy control in dermatology and immunology, which will help to better understand pathomechanisms involving MC accumulation, activation and degranulation and to characterize the effects of therapies that target MCs.
Collapse
Affiliation(s)
- Marius Kröger
- Department of Dermatology, Venerology and Allergology, Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Jörg Scheffel
- Department of Dermatology, Venerology and Allergology, Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Viktor V Nikolaev
- Department of Dermatology, Venerology and Allergology, Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Faculty of Physics, Tomsk State University, Lenin Ave. 36, 634050, Tomsk, Russia
| | - Evgeny A Shirshin
- Faculty of Physics, Lomonosov Moscow State University, Leninskie gory 1/2, 119991, Moscow, Russia
| | - Frank Siebenhaar
- Department of Dermatology, Venerology and Allergology, Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Johannes Schleusener
- Department of Dermatology, Venerology and Allergology, Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Jürgen Lademann
- Department of Dermatology, Venerology and Allergology, Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Marcus Maurer
- Department of Dermatology, Venerology and Allergology, Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Maxim E Darvin
- Department of Dermatology, Venerology and Allergology, Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| |
Collapse
|
28
|
Mnich ME, van Dalen R, van Sorge NM. C-Type Lectin Receptors in Host Defense Against Bacterial Pathogens. Front Cell Infect Microbiol 2020; 10:309. [PMID: 32733813 PMCID: PMC7358460 DOI: 10.3389/fcimb.2020.00309] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 05/22/2020] [Indexed: 12/11/2022] Open
Abstract
Antigen-presenting cells (APCs) are present throughout the human body—in tissues, at barrier sites and in the circulation. They are critical for processing external signals to instruct both local and systemic responses toward immune tolerance or immune defense. APCs express an extensive repertoire of pattern-recognition receptors (PRRs) to detect and transduce these signals. C-type lectin receptors (CLRs) comprise a subfamily of PRRs dedicated to sensing glycans, including those expressed by commensal and pathogenic bacteria. This review summarizes recent findings on the recognition of and responses to bacteria by membrane-expressed CLRs on different APC subsets, which are discussed according to the primary site of infection. Many CLR-bacterial interactions promote bacterial clearance, whereas other interactions are exploited by bacteria to enhance their pathogenic potential. The discrimination between protective and virulence-enhancing interactions is essential to understand which interactions to target with new prophylactic or treatment strategies. CLRs are also densely concentrated at APC dendrites that sample the environment across intact barrier sites. This suggests an–as yet–underappreciated role for CLR-mediated recognition of microbiota-produced glycans in maintaining tolerance at barrier sites. In addition to providing a concise overview of identified CLR-bacteria interactions, we discuss the main challenges and potential solutions for the identification of new CLR-bacterial interactions, including those with commensal bacteria, and for in-depth structure-function studies on CLR-bacterial glycan interactions. Finally, we highlight the necessity for more relevant tissue-specific in vitro, in vivo and ex vivo models to develop therapeutic applications in this area.
Collapse
Affiliation(s)
- Malgorzata E Mnich
- Medical Microbiology, UMC Utrecht, Utrecht University, Utrecht, Netherlands.,GSK, Siena, Italy
| | - Rob van Dalen
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| | - Nina M van Sorge
- Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Netherlands Reference Laboratory for Bacterial Meningitis, Amsterdam University Medical Center, Amsterdam, Netherlands
| |
Collapse
|
29
|
Konieczny P, Naik S. Warp Speed Ahead! Technology-Driven Breakthroughs in Skin Immunity and Inflammatory Disease. J Invest Dermatol 2020; 141:15-18. [PMID: 32533963 DOI: 10.1016/j.jid.2020.05.084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/01/2020] [Accepted: 05/10/2020] [Indexed: 10/24/2022]
Abstract
The skin's physical barrier is reinforced by an arsenal of immune cells that actively patrol the tissue and respond swiftly to penetrating microbes, noxious agents, and injurious stimuli. When unchecked, these same immune cells drive diseases such as psoriasis, atopic dermatitis, and alopecia. Rapidly advancing microscopy, animal modeling, and genomic and computational technologies have illuminated the complexity of the cutaneous immune cells and their functions in maintaining skin health and driving diseases. Here, we discuss the recent technology-driven breakthroughs that have transformed our understanding of skin immunity and highlight burgeoning areas that hold great promise for future discoveries.
Collapse
Affiliation(s)
- Piotr Konieczny
- Department of Pathology, Department of Medicine, and Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, New York, USA
| | - Shruti Naik
- Department of Pathology, Department of Medicine, and Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, New York, USA.
| |
Collapse
|
30
|
Husna N, Gascoigne NRJ, Tey HL, Ng LG, Tan Y. Reprint of "Multi-modal image cytometry approach - From dynamic to whole organ imaging". Cell Immunol 2020; 350:104086. [PMID: 32169249 DOI: 10.1016/j.cellimm.2020.104086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 06/18/2019] [Accepted: 06/18/2019] [Indexed: 12/13/2022]
Abstract
Optical imaging is a valuable tool to visualise biological processes in the context of the tissue. Each imaging modality provides the biologist with different types of information - cell dynamics and migration over time can be tracked with time-lapse imaging (e.g. intra-vital imaging); an overview of whole tissues can be acquired using optical clearing in conjunction with light sheet microscopy; finer details such as cellular morphology and fine nerve tortuosity can be imaged at higher resolution using the confocal microscope. Multi-modal imaging combined with image cytometry - a form of quantitative analysis of image datasets - provides an objective basis for comparing between sample groups. Here, we provide an overview of technical aspects to look out for in an image cytometry workflow, and discuss issues related to sample preparation, image post-processing and analysis for intra-vital and whole organ imaging.
Collapse
Affiliation(s)
- Nazihah Husna
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, 8A Biomedical Grove, Singapore 138648, Singapore; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2, Singapore 117545, Singapore
| | - Nicholas R J Gascoigne
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2, Singapore 117545, Singapore
| | - Hong Liang Tey
- National Skin Centre, 1 Mandalay Road, Singapore 308205, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Singapore 308232, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore 117597, Singapore
| | - Lai Guan Ng
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, 8A Biomedical Grove, Singapore 138648, Singapore; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2, Singapore 117545, Singapore.
| | - Yingrou Tan
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, 8A Biomedical Grove, Singapore 138648, Singapore; National Skin Centre, 1 Mandalay Road, Singapore 308205, Singapore.
| |
Collapse
|
31
|
Husna N, Gascoigne NR, Tey HL, Ng LG, Tan Y. Multi-modal image cytometry approach – From dynamic to whole organ imaging. Cell Immunol 2019; 344:103946. [DOI: 10.1016/j.cellimm.2019.103946] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 06/18/2019] [Accepted: 06/18/2019] [Indexed: 12/27/2022]
|
32
|
Bertram KM, Tong O, Royle C, Turville SG, Nasr N, Cunningham AL, Harman AN. Manipulation of Mononuclear Phagocytes by HIV: Implications for Early Transmission Events. Front Immunol 2019; 10:2263. [PMID: 31616434 PMCID: PMC6768965 DOI: 10.3389/fimmu.2019.02263] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 09/09/2019] [Indexed: 12/11/2022] Open
Abstract
Mononuclear phagocytes are antigen presenting cells that play a key role in linking the innate and adaptive immune systems. In tissue, these consist of Langerhans cells, dendritic cells and macrophages, all of which express the key HIV entry receptors CD4 and CCR5 making them directly infectible with HIV. Mononuclear phagocytes are the first cells of the immune system to interact with invading pathogens such as HIV. Each cell type expresses a specific repertoire of pathogen binding receptors which triggers pathogen uptake and the release of innate immune cytokines. Langerhans cells and dendritic cells migrate to lymph nodes and present antigens to CD4 T cells, whereas macrophages remain tissue resident. Here we review how HIV-1 manipulates these cells by blocking their ability to produce innate immune cytokines and taking advantage of their antigen presenting cell function in order to gain transport to its primary target cells, CD4 T cells.
Collapse
Affiliation(s)
- Kirstie Melissa Bertram
- School of Medical Sciences, University of Sydney, Sydney, NSW, Australia.,Center for Virus Research, The Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Orion Tong
- School of Medical Sciences, University of Sydney, Sydney, NSW, Australia.,Center for Virus Research, The Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Caroline Royle
- School of Medical Sciences, University of Sydney, Sydney, NSW, Australia.,Center for Virus Research, The Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Stuart Grant Turville
- HIV Biology, Kirby Institute, Kensington, NSW, Australia.,The University of New South Whales, Sydney, NSW, Australia
| | - Najla Nasr
- School of Medical Sciences, University of Sydney, Sydney, NSW, Australia.,Center for Virus Research, The Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Anthony Lawrence Cunningham
- School of Medical Sciences, University of Sydney, Sydney, NSW, Australia.,Center for Virus Research, The Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Andrew Nicholas Harman
- School of Medical Sciences, University of Sydney, Sydney, NSW, Australia.,Center for Virus Research, The Westmead Institute for Medical Research, Sydney, NSW, Australia
| |
Collapse
|
33
|
Bertram KM, Botting RA, Baharlou H, Rhodes JW, Rana H, Graham JD, Patrick E, Fletcher J, Plasto TM, Truong NR, Royle C, Doyle CM, Tong O, Nasr N, Barnouti L, Kohout MP, Brooks AJ, Wines MP, Haertsch P, Lim J, Gosselink MP, Ctercteko G, Estes JD, Churchill MJ, Cameron PU, Hunter E, Haniffa MA, Cunningham AL, Harman AN. Identification of HIV transmitting CD11c + human epidermal dendritic cells. Nat Commun 2019; 10:2759. [PMID: 31227717 PMCID: PMC6588576 DOI: 10.1038/s41467-019-10697-w] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 05/24/2019] [Indexed: 11/10/2022] Open
Abstract
Langerhans cells (LC) are thought to be the only mononuclear phagocyte population in the epidermis where they detect pathogens. Here, we show that CD11c+ dendritic cells (DCs) are also present. These cells are transcriptionally similar to dermal cDC2 but are more efficient antigen-presenting cells. Compared to LCs, epidermal CD11c+ DCs are enriched in anogenital tissues where they preferentially interact with HIV, express the higher levels of HIV entry receptor CCR5, support the higher levels of HIV uptake and replication and are more efficient at transmitting the virus to CD4 T cells. Importantly, these findings are observed using both a lab-adapted and transmitted/founder strain of HIV. We also describe a CD33low cell population, which is transcriptionally similar to LCs but does not appear to function as antigen-presenting cells or acts as HIV target cells. Our findings reveal that epidermal DCs in anogenital tissues potentially play a key role in sexual transmission of HIV. Composition and function of immune populations at barrier surfaces is crucial for response to infection. Here, the authors identify a population of dendritic cells in human epidermis, abundant in anogenital epithelia and distinct from Langerhans cells by surface phenotype and by high capacity for HIV infection and transmission.
Collapse
Affiliation(s)
- Kirstie M Bertram
- Centre for Virus Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, New South Wales, 2145, Australia.,The University of Sydney, Sydney, 2006, New South Wales, Australia
| | - Rachel A Botting
- Centre for Virus Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, New South Wales, 2145, Australia.,The University of Sydney, Sydney, 2006, New South Wales, Australia.,Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Heeva Baharlou
- Centre for Virus Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, New South Wales, 2145, Australia.,The University of Sydney, Sydney, 2006, New South Wales, Australia
| | - Jake W Rhodes
- Centre for Virus Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, New South Wales, 2145, Australia.,The University of Sydney, Sydney, 2006, New South Wales, Australia
| | - Hafsa Rana
- Centre for Virus Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, New South Wales, 2145, Australia.,The University of Sydney, Sydney, 2006, New South Wales, Australia
| | - J Dinny Graham
- Centre for Virus Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, New South Wales, 2145, Australia.,The University of Sydney, Sydney, 2006, New South Wales, Australia
| | - Ellis Patrick
- Centre for Virus Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, New South Wales, 2145, Australia.,The University of Sydney, Sydney, 2006, New South Wales, Australia
| | - James Fletcher
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Toby M Plasto
- Centre for Virus Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, New South Wales, 2145, Australia.,The University of Sydney, Sydney, 2006, New South Wales, Australia
| | - Naomi R Truong
- Centre for Virus Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, New South Wales, 2145, Australia.,The University of Sydney, Sydney, 2006, New South Wales, Australia
| | - Caroline Royle
- Centre for Virus Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, New South Wales, 2145, Australia.,The University of Sydney, Sydney, 2006, New South Wales, Australia
| | - Chloe M Doyle
- Centre for Virus Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, New South Wales, 2145, Australia.,The University of Sydney, Sydney, 2006, New South Wales, Australia
| | - Orion Tong
- Centre for Virus Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, New South Wales, 2145, Australia.,The University of Sydney, Sydney, 2006, New South Wales, Australia
| | - Najla Nasr
- Centre for Virus Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, New South Wales, 2145, Australia.,The University of Sydney, Sydney, 2006, New South Wales, Australia
| | - Laith Barnouti
- Australia Plastic Surgery, 185-211, Broadway, Sydney, New South Wales, 2007, Australia
| | - Mark P Kohout
- Australia Plastic Surgery, 185-211, Broadway, Sydney, New South Wales, 2007, Australia
| | - Andrew J Brooks
- Westmead Hospital, Westmead, New South Wales, 2145, Australia
| | - Michael P Wines
- Royal North Shore Hospital, Reserve Rd, St Leonards, New South Wales, 2065, Australia
| | - Peter Haertsch
- Burns Unit, Concord Repatriation General Hospital, Sydney, 2139, New South Wales, Australia
| | - Jake Lim
- Dr Jake Lim PLC, Shop 12, Cnr of Aird & Marsden Street, Parramatta, New South Wales, 2150, Australia
| | - Martijn P Gosselink
- Centre for Virus Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, New South Wales, 2145, Australia.,Westmead Hospital, Westmead, New South Wales, 2145, Australia
| | - Grahame Ctercteko
- Centre for Virus Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, New South Wales, 2145, Australia.,Westmead Hospital, Westmead, New South Wales, 2145, Australia
| | - Jacob D Estes
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc, Frederick, MD, 21702, USA
| | - Melissa J Churchill
- School of Health and Biomedical Sciences, College of Science, Engineering and Health, RMIT University, Melbourne, Victoria, 3001, Australia
| | - Paul U Cameron
- The Peter Doherty Institute for Infection and Immunity, 792 Elizabeth Street, Melbourne, Victoria, 3000, Australia
| | - Eric Hunter
- Emory Vaccine Center, 954 Gatewood Road, Atlanta, GA, 30329, USA
| | - Muzlifah A Haniffa
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.,Department of Dermatology, Royal Victoria Infirmary, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, NE2 4LP, UK
| | - Anthony L Cunningham
- Centre for Virus Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, New South Wales, 2145, Australia.,The University of Sydney, Sydney, 2006, New South Wales, Australia
| | - Andrew N Harman
- Centre for Virus Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, New South Wales, 2145, Australia. .,The University of Sydney, Sydney, 2006, New South Wales, Australia.
| |
Collapse
|
34
|
Rhodes JW, Tong O, Harman AN, Turville SG. Human Dendritic Cell Subsets, Ontogeny, and Impact on HIV Infection. Front Immunol 2019; 10:1088. [PMID: 31156637 PMCID: PMC6532592 DOI: 10.3389/fimmu.2019.01088] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 04/29/2019] [Indexed: 12/18/2022] Open
Abstract
Dendritic cells (DCs) play important roles in orchestrating host immunity against invading pathogens, representing one of the first responders to infection by mucosal invaders. From their discovery by Ralph Steinman in the 1970s followed shortly after with descriptions of their in vivo diversity and distribution by Derek Hart, we are still continuing to progressively elucidate the spectrum of DCs present in various anatomical compartments. With the power of high-dimensional approaches such as single-cell sequencing and multiparameter cytometry, recent studies have shed new light on the identities and functions of DC subtypes. Notable examples include the reclassification of plasmacytoid DCs as purely interferon-producing cells and re-evaluation of intestinal conventional DCs and macrophages as derived from monocyte precursors. Collectively, these observations have changed how we view these cells not only in steady-state immunity but also during disease and infection. In this review, we will discuss the current landscape of DCs and their ontogeny, and how this influences our understanding of their roles during HIV infection.
Collapse
Affiliation(s)
- Jake William Rhodes
- Centre for Virus Research, The Westmead Institute for Medical Research, Sydney, NSW, Australia.,Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Orion Tong
- Centre for Virus Research, The Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Andrew Nicholas Harman
- Centre for Virus Research, The Westmead Institute for Medical Research, Sydney, NSW, Australia.,Discipline of Applied Medical Sciences, School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
| | - Stuart Grant Turville
- University of New South Wales, Sydney, NSW, Australia.,Kirby Institute, Kensington, NSW, Australia
| |
Collapse
|
35
|
Affiliation(s)
- Martijn Pieter Gosselink
- Department of Colorectal Surgery, Westmead Hospital, The University of Sydney Westmead Clinical School, Sydney, NSW, Australia.,Centre for Virus Research, Westmead Institute for Medical Research, Westmead, NSW, Australia.,University of Sydney, Sydney, NSW, Australia
| | - Andrew N Harman
- Centre for Virus Research, Westmead Institute for Medical Research, Westmead, NSW, Australia.,University of Sydney, Sydney, NSW, Australia.,The School of Medical Sciences, University of Sydney, Sydney, Australia
| | - Grahame Ctercteko
- Department of Colorectal Surgery, Westmead Hospital, The University of Sydney Westmead Clinical School, Sydney, NSW, Australia
| |
Collapse
|
36
|
The contraceptive medroxyprogesterone acetate, unlike norethisterone, directly increases R5 HIV-1 infection in human cervical explant tissue at physiologically relevant concentrations. Sci Rep 2019; 9:4334. [PMID: 30867477 PMCID: PMC6416361 DOI: 10.1038/s41598-019-40756-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 02/19/2019] [Indexed: 02/06/2023] Open
Abstract
The intramuscular progestin-only injectable contraceptive, depo-medroxyprogesterone acetate (DMPA-IM), is more widely used in Sub-Saharan Africa than another injectable contraceptive, norethisterone enanthate (NET-EN). Epidemiological data show a significant 1.4-fold increased risk of HIV-1 acquisition for DMPA-IM usage, while no such association is shown from limited data for NET-EN. We show that MPA, unlike NET, significantly increases R5-tropic but not X4-tropic HIV-1 replication ex vivo in human endocervical and ectocervical explant tissue from pre-menopausal donors, at physiologically relevant doses. Results support a mechanism whereby MPA, unlike NET, acts via the glucocorticoid receptor (GR) to increase HIV-1 replication in cervical tissue by increasing the relative frequency of CD4+ T cells and activated monocytes. We show that MPA, unlike NET, increases mRNA expression of the CD4 HIV-1 receptor and CCR5 but not CXCR4 chemokine receptors, via the GR. However, increased density of CD4 on CD3+ cells was not observed with MPA by flow cytometry of digested tissue. Results suggest that DMPA-IM may increase HIV-1 acquisition in vivo at least in part via direct effects on cervical tissue to increase founder R5-tropic HIV-1 replication. Our findings support differential biological mechanisms and disaggregation of DMPA-IM and NET-EN regarding HIV-1 acquisition risk category for use in high risk areas.
Collapse
|
37
|
Rogers KJ, Maury W. The role of mononuclear phagocytes in Ebola virus infection. J Leukoc Biol 2018; 104:717-727. [PMID: 30095866 DOI: 10.1002/jlb.4ri0518-183r] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 07/09/2018] [Accepted: 07/12/2018] [Indexed: 12/17/2022] Open
Abstract
The filovirus, Zaire Ebolavirus (EBOV), infects tissue macrophages (Mϕs) and dendritic cells (DCs) early during infection. Viral infection of both cells types is highly productive, leading to increased viral load. However, virus infection of these two cell types results in different consequences for cellular function. Infection of Mϕs stimulates the production of proinflammatory and immunomodulatory cytokines and chemokines, leading to the production of a cytokine storm, while simultaneously increasing tissue factor production and thus facilitating disseminated intravascular coagulation. In contrast, EBOV infection of DCs blocks DC maturation and antigen presentation rendering these cells unable to communicate with adaptive immune response elements. Details of the known interactions of these cells with EBOV are reviewed here. We also identify a number of unanswered questions that remain about interactions of filoviruses with these cells.
Collapse
Affiliation(s)
- Kai J Rogers
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
| | - Wendy Maury
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|