1
|
Arshad S, Rana H, Truong NR, Pattamatta U, Bertram KM, White A, Chinnery HR, Carnt NA, Cunningham AL. Herpes simplex virus type-1 infection and spread in a novel porcine corneal explant model is restricted to the epithelium. PLoS Pathog 2025; 21:e1013162. [PMID: 40315239 PMCID: PMC12068712 DOI: 10.1371/journal.ppat.1013162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 05/12/2025] [Accepted: 04/24/2025] [Indexed: 05/04/2025] Open
Abstract
Herpes Keratitis (HK) is a debilitating infection of the cornea that remains the leading cause of infectious blindness in developed countries. Caused primarily by herpes simplex virus type 1 (HSV-1), it is associated with recurrent inflammation, leading to corneal scarring. This study investigated the initial events during acute HSV-1 infection in the cornea by adapting our human anogenital mucosal explant model to a HSV-1 infected porcine corneal explant model. We infected these corneas topically via high-density microarray patches (HD-MAPs) dipped in GFP-labelled HSV-1. Virus infection and spread was detected by both GFP protein and RNAscope, adapted for HSV-1 DNA. The punctures were consistent, usually in the epithelium but some extended into the underlying stroma. However, HSV-1 was restricted to the corneal epithelium, without spread through the anterior limiting membrane (ALM) or Bowman's layer into the stroma nor to the uppermost epithelial layer. This layer expressed SPRR1A similarly to the stratum granulosum of skin which is refractory to HSV-1 infection. In corneas where infected epithelial cells extended to the ALM, SPRR1A was also observed in this layer, suggesting it may contribute to its barrier function. Such studies of HSV-1 infection and spread will help improve therapy for HK and vaccine design to prevent it.
Collapse
Affiliation(s)
- Sana Arshad
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Centre for Vision Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Hafsa Rana
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Naomi R. Truong
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Ushasree Pattamatta
- Centre for Vision Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, 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, Sydney, New South Wales, Australia
| | - Andrew White
- Centre for Vision Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Holly R. Chinnery
- Department of Optometry and Vision Science, The University of Western Australia, Crawley, Western Australia, Australia
| | - Nicole A. Carnt
- Centre for Vision Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- School of Optometry and Vision Science, University of New South Wales, Kensington, New South Wales, Australia
- Institute of Ophthalmology, University College London, London, United Kingdom
| | - Anthony L. Cunningham
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
2
|
Kotlyar J, Granstein RD. Neuroimmunology of psoriasis: Possible roles for calcitonin gene-related peptide in its pathogenesis. Brain Behav Immun Health 2025; 44:100958. [PMID: 40008232 PMCID: PMC11851231 DOI: 10.1016/j.bbih.2025.100958] [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: 09/24/2024] [Revised: 01/21/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025] Open
Abstract
The nervous system has a complex interplay with the immune system, especially at barrier sites such as the skin. This allows it to play a role in a variety of cutaneous inflammatory disorders such as psoriasis, exerting effects on various immune cells via effector molecules such as neuropeptides. In this review, we discuss the role of calcitonin gene-related peptide in modulating the immune system and inflammation, with a focus on psoriasis.
Collapse
Affiliation(s)
- Joshua Kotlyar
- Israel Englander Department of Dermatology, Weill Cornell Medicine, 1305 York Avenue, WGC9, New York, NY, 10021, USA
- SUNY Downstate Health Sciences University College of Medicine, 450 Clarkson Avenue, Brooklyn, NY, 11203, USA
| | - Richard D. Granstein
- Israel Englander Department of Dermatology, Weill Cornell Medicine, 1305 York Avenue, WGC9, New York, NY, 10021, USA
| |
Collapse
|
3
|
Warner van Dijk FA, Bertram KM, O’Neil TR, Li Y, Buffa DJ, Harman AN, Cunningham AL, Nasr N. Recent Advances in Our Understanding of Human Inflammatory Dendritic Cells in Human Immunodeficiency Virus Infection. Viruses 2025; 17:105. [PMID: 39861894 PMCID: PMC11768623 DOI: 10.3390/v17010105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 01/03/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
Anogenital inflammation is a critical risk factor for HIV acquisition. The primary preventative HIV intervention, pre-exposure prophylaxis (PrEP), is ineffective in blocking transmission in anogenital inflammation. Pre-existing sexually transmitted diseases (STIs) and anogenital microbiota dysbiosis are the leading causes of inflammation, where inflammation is extensive and often asymptomatic and undiagnosed. Dendritic cells (DCs), as potent antigen-presenting cells, are among the first to capture HIV upon its entry into the mucosa, and they subsequently transport the virus to CD4 T cells, the primary HIV target cells. This increased HIV susceptibility in inflamed tissue likely stems from a disrupted epithelial barrier integrity, phenotypic changes in resident DCs and an influx of inflammatory HIV target cells, including DCs and CD4 T cells. Gaining insight into how HIV interacts with specific inflammatory DC subsets could inform the development of new therapeutic strategies to block HIV transmission. However, little is known about the early stages of HIV capture and transmission in inflammatory environments. Here, we review the currently characterised inflammatory-tissue DCs and their interactions with HIV.
Collapse
Affiliation(s)
- Freja A. Warner van Dijk
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead 2145, Australia; (F.A.W.v.D.); (K.M.B.); (T.R.O.); (Y.L.); (D.J.B.); (A.N.H.)
- Faculty of Medicine and Health, Sydney Infectious Diseases Institute, School of Medical Sciences, The University of Sydney, Sydney 2006, Australia
| | - Kirstie M. Bertram
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead 2145, Australia; (F.A.W.v.D.); (K.M.B.); (T.R.O.); (Y.L.); (D.J.B.); (A.N.H.)
- Faculty of Medicine and Health, Sydney Infectious Diseases Institute, School of Medical Sciences, The University of Sydney, Sydney 2006, Australia
| | - Thomas R. O’Neil
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead 2145, Australia; (F.A.W.v.D.); (K.M.B.); (T.R.O.); (Y.L.); (D.J.B.); (A.N.H.)
- Faculty of Medicine and Health, Sydney Infectious Diseases Institute, School of Medical Sciences, The University of Sydney, Sydney 2006, Australia
| | - Yuchen Li
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead 2145, Australia; (F.A.W.v.D.); (K.M.B.); (T.R.O.); (Y.L.); (D.J.B.); (A.N.H.)
- Faculty of Medicine and Health, Sydney Infectious Diseases Institute, School of Medical Sciences, The University of Sydney, Sydney 2006, Australia
| | - Daniel J. Buffa
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead 2145, Australia; (F.A.W.v.D.); (K.M.B.); (T.R.O.); (Y.L.); (D.J.B.); (A.N.H.)
- Faculty of Medicine and Health, Sydney Infectious Diseases Institute, School of Medical Sciences, The University of Sydney, Sydney 2006, Australia
| | - Andrew N. Harman
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead 2145, Australia; (F.A.W.v.D.); (K.M.B.); (T.R.O.); (Y.L.); (D.J.B.); (A.N.H.)
- Faculty of Medicine and Health, Sydney Infectious Diseases Institute, School of Medical Sciences, The University of Sydney, Sydney 2006, Australia
| | - Anthony L. Cunningham
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead 2145, Australia; (F.A.W.v.D.); (K.M.B.); (T.R.O.); (Y.L.); (D.J.B.); (A.N.H.)
- Faculty of Medicine and Health, Sydney Infectious Diseases Institute, School of Medical Sciences, The University of Sydney, Sydney 2006, Australia
| | - Najla Nasr
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead 2145, Australia; (F.A.W.v.D.); (K.M.B.); (T.R.O.); (Y.L.); (D.J.B.); (A.N.H.)
- Faculty of Medicine and Health, Sydney Infectious Diseases Institute, School of Medical Sciences, The University of Sydney, Sydney 2006, Australia
| |
Collapse
|
4
|
Laguía F, Chojnacki J, Erkizia I, Geli MI, Enrich C, Martinez-Picado J, Resa-Infante P. Massive endocytosis mechanisms are involved in uptake of HIV-1 particles by monocyte-derived dendritic cells. Front Immunol 2025; 15:1505840. [PMID: 39867902 PMCID: PMC11757119 DOI: 10.3389/fimmu.2024.1505840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 12/11/2024] [Indexed: 01/28/2025] Open
Abstract
Introduction HIV-1 exploits dendritic cells (DCs) to spread throughout the body via specific recognition of gangliosides present on the viral envelope by the CD169/Siglec-1 membrane receptor. This interaction triggers the internalization of HIV-1 within a structure known as the sac-like compartment. While the mechanism underlying sac-like compartment formation remains elusive, prior research indicates that the process is clathrin-independent and cell membrane cholesterol-dependent and involves transient disruption of cortical actin. Here, we investigate the potential involvement of massive endocytosis (MEND) in this process. Methods We used live cell confocal imaging to measure the dimensions and dynamics of the compartment. We assessed the role of actin and cholesterol in fixed and live cells using confocal microscopy and evaluated the effect of PI3K and protein palmytoilation inhibitors during viral uptake. Results Our data demonstrate extensive plasma membrane invagination based on sac-like compartment dimensions (2.9 μm in diameter and 20 μm3 in volume). We showed that the cholesterol concentration doubles within the regions of viral uptake, suggesting lipid-phase separation, and that development of the sac-like compartment is accompanied by transient depolarization of cortical actin. Moreover, we observed that protein palmitoylation and PI3K inhibition reduce the sac-like compartment formation rate from 70% to 20% and 40%, respectively. Conclusions Our results indicate the involvement of MEND mechanisms during sac-like compartment formation.
Collapse
Affiliation(s)
| | - Jakub Chojnacki
- IrsiCaixa, Badalona, Spain
- CIBERINFEC, Madrid, Spain
- Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
| | | | - María Isabel Geli
- Department of Cell Biology, Institute for Molecular Biology of Barcelona (IBMB, CSIC), Barcelona, Spain
| | - Carlos Enrich
- Cell Compartments and Signaling Group, Institut d’Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Javier Martinez-Picado
- IrsiCaixa, Badalona, Spain
- CIBERINFEC, Madrid, Spain
- Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
- University of Vic-Central University of Catalonia (UVic-UCC), Vic, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Patricia Resa-Infante
- IrsiCaixa, Badalona, Spain
- CIBERINFEC, Madrid, Spain
- Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
- University of Vic-Central University of Catalonia (UVic-UCC), Vic, Spain
| |
Collapse
|
5
|
Hu K, O’Neil TR, Baharlou H, Austin PJ, Karrasch JF, Sarkawt L, Li Y, Bertram KM, Cunningham AL, Patrick E, Harman AN. The spatial biology of HIV infection. PLoS Pathog 2025; 21:e1012888. [PMID: 39854613 PMCID: PMC11760614 DOI: 10.1371/journal.ppat.1012888] [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] [Indexed: 01/26/2025] Open
Abstract
HIV infection implicates a spectrum of tissues in the human body starting with viral transmission in the anogenital tract and subsequently persisting in lymphoid tissues and brain. Though studies using isolated cells have contributed significantly towards our understanding of HIV infection, the tissue microenvironment is characterised by a complex interplay of a range of factors, all of which can influence the course of infection but are otherwise missed in ex vivo studies. To address this knowledge gap, it is necessary to investigate the dynamics of infection and the host immune response in situ using imaging-based approaches. Over the last decade, emerging imaging techniques have continually redefined the limits of detection, both in terms of the scope and the scale of the targets. In doing so, this has opened up new questions that can be answered by in situ studies. This review discusses the high-dimensional imaging modalities that are now available and their application towards understanding the spatial biology of HIV infection.
Collapse
Affiliation(s)
- Kevin Hu
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Thomas R. O’Neil
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Heeva Baharlou
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Paul J. Austin
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Brain and Mind Centre, School of Medical of Medical Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - Jackson F. Karrasch
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Brain and Mind Centre, School of Medical of Medical Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - Lara Sarkawt
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Yuchen Li
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Kirstie M. Bertram
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Anthony L. Cunningham
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Ellis Patrick
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- School of Mathematics and Statistics, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Andrew N. Harman
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
6
|
Rana H, Truong NR, Sirimanne DR, Cunningham AL. Breaching the Barrier: Investigating Initial Herpes Simplex Viral Infection and Spread in Human Skin and Mucosa. Viruses 2024; 16:1790. [PMID: 39599904 PMCID: PMC11599041 DOI: 10.3390/v16111790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/13/2024] [Accepted: 11/16/2024] [Indexed: 11/29/2024] Open
Abstract
Herpes simplex virus (HSV) is sexually transmitted via the anogenital mucosa where it initially infects epidermal keratinocytes and mononuclear phagocytes (MNPs). It then spreads to the dorsal root ganglion via sensory nerve endings, to remain latent for life with periodic reactivation. Currently, there is no cure or vaccine. Initial or recurrent HSV infection can produce serious complications and mediate acquisition of HIV. This review outlines the initial events after the HSV infection of human anogenital mucosa to determine the optimal window to target the virus before it becomes latent. After infection, HSV spreads rapidly within the mid-layers of epidermal keratinocytes in the explanted human inner foreskin. Infected cells produce chemokines, which modulate nectin-1 distribution on the surface of adjacent keratinocytes, facilitating viral spread. Epidermal Langerhans cells and dendritic cells become infected with HSV followed by a "viral relay" to dermal MNPs, which then present viral antigen to T cells in the dermis or lymph nodes. These data indicate the need for interruption of spread within 24 h by diffusible vaccine-induced mediators such as antiviral cytokines from resident immune cells or antibodies. Intradermal/mucosal vaccines would need to target the relevant dermal MNPs to induce HSV-specific CD4+ and CD8+ T cells.
Collapse
Affiliation(s)
- Hafsa Rana
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW 2145, Australia; (H.R.); (N.R.T.); (D.R.S.)
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Naomi R. Truong
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW 2145, Australia; (H.R.); (N.R.T.); (D.R.S.)
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Dona R. Sirimanne
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW 2145, Australia; (H.R.); (N.R.T.); (D.R.S.)
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Anthony L. Cunningham
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW 2145, Australia; (H.R.); (N.R.T.); (D.R.S.)
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
7
|
Haga K, Tokui T, Miyamoto K, Takai‐Todaka R, Kudo S, Ishikawa A, Ishiyama R, Kato A, Yokoyama M, Katayama K, Nakanishi A. Neonatal Fc receptor is a functional receptor for classical human astrovirus. Genes Cells 2024; 29:983-1001. [PMID: 39266307 PMCID: PMC11555631 DOI: 10.1111/gtc.13160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 08/24/2024] [Indexed: 09/14/2024]
Abstract
Human astrovirus (HAstV) is a global cause of gastroenteritis in infants, the elderly, and the immunocompromised. However, the molecular mechanisms that control its susceptibility are not fully understood, as the functional receptor used by the virus has yet to be identified. Here, a genome-wide CRISPR-Cas9 library screen in Caco2 cells revealed that the neonatal Fc receptor (FcRn) can function as a receptor for classical HAstV (Mamastrovirus genotype 1). Deletion of FCGRT or B2M, which encode subunits of FcRn, rendered Caco2 cells and intestinal organoid cells resistant to HAstV infection. We also showed that human FcRn expression renders non-susceptible cells permissive to viral infection and that FcRn binds directly to the HAstV spike protein. Therefore, our findings provide insight into the entry mechanism of HAstV into susceptible cells. We anticipate that this information can be used to develop new therapies targeting human astroviruses, providing new strategies to treat this global health issue.
Collapse
Affiliation(s)
- Kei Haga
- Laboratory of Viral Infection Control, Department of Infection Control and Immunology, Ōmura Satoshi Memorial Institute & Graduate School of Infection Control SciencesKitasato UniversityTokyoJapan
| | - Takashi Tokui
- Laboratory of Viral Infection Control, Department of Infection Control and Immunology, Ōmura Satoshi Memorial Institute & Graduate School of Infection Control SciencesKitasato UniversityTokyoJapan
| | - Kana Miyamoto
- Laboratory of Viral Infection Control, Department of Infection Control and Immunology, Ōmura Satoshi Memorial Institute & Graduate School of Infection Control SciencesKitasato UniversityTokyoJapan
| | - Reiko Takai‐Todaka
- Laboratory of Viral Infection Control, Department of Infection Control and Immunology, Ōmura Satoshi Memorial Institute & Graduate School of Infection Control SciencesKitasato UniversityTokyoJapan
| | - Shiori Kudo
- Laboratory of Viral Infection Control, Department of Infection Control and Immunology, Ōmura Satoshi Memorial Institute & Graduate School of Infection Control SciencesKitasato UniversityTokyoJapan
| | - Azusa Ishikawa
- Laboratory of Viral Infection Control, Department of Infection Control and Immunology, Ōmura Satoshi Memorial Institute & Graduate School of Infection Control SciencesKitasato UniversityTokyoJapan
| | - Ryoka Ishiyama
- Laboratory of Viral Infection Control, Department of Infection Control and Immunology, Ōmura Satoshi Memorial Institute & Graduate School of Infection Control SciencesKitasato UniversityTokyoJapan
| | - Akiko Kato
- National Center for Geriatrics and Gerontology, Department of Aging InterventionLaboratory of Gene Therapy, and Laboratory for Radiation safetyAichiJapan
| | - Masaru Yokoyama
- Pathogen Genomics Center, National Institute of Infectious DiseasesTokyoJapan
| | - Kazuhiko Katayama
- Laboratory of Viral Infection Control, Department of Infection Control and Immunology, Ōmura Satoshi Memorial Institute & Graduate School of Infection Control SciencesKitasato UniversityTokyoJapan
| | - Akira Nakanishi
- National Center for Geriatrics and Gerontology, Department of Aging InterventionLaboratory of Gene Therapy, and Laboratory for Radiation safetyAichiJapan
- Department of Biology‐Oriented Science and TechnologyKindai UniversityWakayamaJapan
| |
Collapse
|
8
|
Parthasarathy S, Moreno de Lara L, Carrillo-Salinas FJ, Werner A, Borchers A, Iyer V, Vogell A, Fortier JM, Wira CR, Rodriguez-Garcia M. Human genital dendritic cell heterogeneity confers differential rapid response to HIV-1 exposure. Front Immunol 2024; 15:1472656. [PMID: 39524443 PMCID: PMC11543421 DOI: 10.3389/fimmu.2024.1472656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024] Open
Abstract
Dendritic cells (DCs) play critical roles in HIV pathogenesis and require further investigation in the female genital tract, a main portal of entry for HIV infection. Here we characterized genital DC populations at the single cell level and how DC subsets respond to HIV immediately following exposure. We found that the genital CD11c+HLA-DR+ myeloid population contains three DC subsets (CD1c+ DC2s, CD14+ monocyte-derived DCs and CD14+CD1c+ DC3s) and two monocyte/macrophage populations with distinct functional and phenotypic properties during homeostasis. Following HIV exposure, the antiviral response was dominated by DCs' rapid secretory response, activation of non-classical inflammatory pathways and host restriction factors. Further, we uncovered subset-specific differences in anti-HIV responses. CD14+ DCs were the main population activated by HIV and mediated the secretory antimicrobial response, while CD1c+ DC2s activated inflammasome pathways and IFN responses. Identification of subset-specific responses to HIV immediately after exposure could aid targeted strategies to prevent HIV infection.
Collapse
Affiliation(s)
- Siddharth Parthasarathy
- Department of Immunology, Tufts University School of Medicine, Boston, MA, United States
- Immunology Graduate Program, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States
| | - Laura Moreno de Lara
- Department of Immunology, Tufts University School of Medicine, Boston, MA, United States
| | | | - Alexandra Werner
- Department of Immunology, Tufts University School of Medicine, Boston, MA, United States
- Immunology Graduate Program, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States
- C.S Mott Center for Human Growth and Development, Department of Obstetrics & Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Anna Borchers
- Department of Immunology, Tufts University School of Medicine, Boston, MA, United States
| | - Vidya Iyer
- Department of Gynecology and Obstetrics, Tufts Medical Center, Boston, MA, United States
- Mass General Research Institute (MGRI), Division of Clinical Research, Massachusetts General Hospital, Boston, MA, United States
| | - Alison Vogell
- Department of Gynecology and Obstetrics, Tufts Medical Center, Boston, MA, United States
| | - Jared M. Fortier
- C.S Mott Center for Human Growth and Development, Department of Obstetrics & Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Charles R. Wira
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Marta Rodriguez-Garcia
- Department of Immunology, Tufts University School of Medicine, Boston, MA, United States
- Immunology Graduate Program, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States
- C.S Mott Center for Human Growth and Development, Department of Obstetrics & Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI, United States
| |
Collapse
|
9
|
Zhu R, Yao X, Li W. Langerhans cells and skin immune diseases. Eur J Immunol 2024; 54:e2250280. [PMID: 39030782 DOI: 10.1002/eji.202250280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 07/04/2024] [Accepted: 07/08/2024] [Indexed: 07/22/2024]
Abstract
Langerhans cells (LCs) are the key antigen-presenting cells in the epidermis in normal conditions and respond differentially to environmental and/or endogenous stimuli, exerting either proinflammatory or anti-inflammatory effects. Current knowledge about LCs mainly originates from studies utilizing mouse models, whereas with the development of single-cell techniques, there has been significant progress for human LCs, which has updated our understanding of the phenotype, ontogeny, differentiation regulation, and function of LCs. In this review, we delineated the progress of human LCs and summarized LCs' function in inflammatory skin diseases, providing new ideas for precise regulation of LC function in the prevention and treatment of skin diseases.
Collapse
Affiliation(s)
- Ronghui Zhu
- Department of Dermatology, Shanghai Institute of Dermatology, Huashan Hospital, Fudan University, Shanghai, P. R. China
- Department of Dermatology, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
- Hubei Province & Key Laboratory of Skin Infection and Immunity, Wuhan, P. R. China
| | - Xu Yao
- Department, of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, P. R. China
| | - Wei Li
- Department of Dermatology, Shanghai Institute of Dermatology, Huashan Hospital, Fudan University, Shanghai, P. R. China
| |
Collapse
|
10
|
Rana H, Truong NR, Johnson B, Baharlou H, Herbert JJ, Kandasamy S, Goddard R, Cohen RC, Wines M, Nasr N, Harman AN, Bertram KM, Sandgren KJ, Cunningham AL. Herpes simplex virus spreads rapidly in human foreskin, partly driven by chemokine-induced redistribution of Nectin-1 on keratinocytes. PLoS Pathog 2024; 20:e1012267. [PMID: 38857290 PMCID: PMC11164381 DOI: 10.1371/journal.ppat.1012267] [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: 11/22/2023] [Accepted: 05/17/2024] [Indexed: 06/12/2024] Open
Abstract
HSV infects keratinocytes in the epidermis of skin via nectin-1. We established a human foreskin explant infection model to investigate HSV entry and spread. HSV1 entry could only be achieved by the topical application of virus via high density microarray projections (HD-MAPs) to the epidermis, which penetrated beyond one third of its thickness, simulating in vivo microtrauma. Rapid lateral spread of HSV1 to a mean of 13 keratinocytes wide occurred after 24 hours and free virus particles were observed between keratinocytes, consistent with an intercellular route of spread. Nectin-1 staining was markedly decreased in foci of infection in the epidermis and in the human keratinocyte HaCaT cell line. Nectin-1 was redistributed, at the protein level, in adjacent uninfected cells surrounding infection, inducible by CCL3, IL-8 (or CXCL8), and possibly CXCL10 and IL-6, thus facilitating spread. These findings provide the first insights into HSV1 entry and spread in human inner foreskin in situ.
Collapse
Affiliation(s)
- Hafsa Rana
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Naomi R. Truong
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Blake Johnson
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Heeva Baharlou
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Jason J. Herbert
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | | | - Robert Goddard
- Research and Development, Vaxxas Pty Ltd., Brisbane, Queensland, Australia
| | - Ralph C. Cohen
- University of Sydney and Australian National University, Children’s Hospital at Westmead, New South Wales, Australia
| | - Michael Wines
- Urology, Sydney Adventist Hospital, Wahroonga, New South Wales, Australia
| | - Najla Nasr
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Andrew N. Harman
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Kirstie M. Bertram
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Kerrie J. Sandgren
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Anthony L. Cunningham
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
11
|
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
|
12
|
O'Brien JA, Karrasch JF, Huang Y, Vine EE, Cunningham AL, Harman AN, Austin PJ. Nerve-myeloid cell interactions in persistent human pain: a reappraisal using updated cell subset classifications. Pain 2024; 165:753-771. [PMID: 37975868 DOI: 10.1097/j.pain.0000000000003106] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 09/04/2023] [Indexed: 11/19/2023]
Abstract
ABSTRACT The past 20 years have seen a dramatic shift in our understanding of the role of the immune system in initiating and maintaining pain. Myeloid cells, including macrophages, dendritic cells, Langerhans cells, and mast cells, are increasingly implicated in bidirectional interactions with nerve fibres in rodent pain models. However, our understanding of the human setting is still poor. High-dimensional functional analyses have substantially changed myeloid cell classifications, with recently described subsets such as epidermal dendritic cells and DC3s unveiling new insight into how myeloid cells interact with nerve fibres. However, it is unclear whether this new understanding has informed the study of human chronic pain. In this article, we perform a scoping review investigating neuroimmune interactions between myeloid cells and peripheral nerve fibres in human chronic pain conditions. We found 37 papers from multiple pain states addressing this aim in skin, cornea, peripheral nerve, endometrium, and tumour, with macrophages, Langerhans cells, and mast cells the most investigated. The directionality of results between studies was inconsistent, although the clearest pattern was an increase in macrophage frequency across conditions, phases, and tissues. Myeloid cell definitions were often outdated and lacked correspondence with the stated cell types of interest; overreliance on morphology and traditional structural markers gave limited insight into the functional characteristics of investigated cells. We therefore critically reappraise the existing literature considering contemporary myeloid cell biology and advocate for the application of established and emerging high-dimensional proteomic and transcriptomic single-cell technologies to clarify the role of specific neuroimmune interactions in chronic pain.
Collapse
Affiliation(s)
- Jayden A O'Brien
- Brain and Mind Centre, The University of Sydney, Sydney, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Jackson F Karrasch
- Brain and Mind Centre, The University of Sydney, Sydney, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, Australia
| | - Yun Huang
- Brain and Mind Centre, The University of Sydney, Sydney, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Erica E Vine
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, Australia
| | - Anthony L Cunningham
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, Australia
| | - Andrew N Harman
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, Australia
| | - Paul J Austin
- Brain and Mind Centre, The University of Sydney, Sydney, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, Australia
| |
Collapse
|
13
|
Lopes KDP, Yu L, Shen X, Qiu Y, Tasaki S, Iatrou A, Beeri MS, Seyfried NT, Menon V, Wang Y, Schneider JA, Cantor H, Bennett DA. Associations of cortical SPP1 and ITGAX with cognition and common neuropathologies in older adults. Alzheimers Dement 2024; 20:525-537. [PMID: 37727065 PMCID: PMC10841499 DOI: 10.1002/alz.13474] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/15/2023] [Accepted: 08/21/2023] [Indexed: 09/21/2023]
Abstract
INTRODUCTION The secreted phosphoprotein 1 (SPP1) gene expressed by CD11c+ cells is known to be associated with microglia activation and neuroinflammatory diseases. As most studies rely on mouse models, we investigated these genes and proteins in the cortical brain tissue of older adults and their role in Alzheimer's disease (AD) and related disorders. METHODS We leveraged protein measurements, single-nuclei, and RNASeq data from the Religious Orders Study and Rush Memory and Aging Project (ROSMAP) of over 1200 samples for association analysis. RESULTS Expression of SPP1 and its encoded protein osteopontin were associated with faster cognitive decline and greater odds of common neuropathologies. At single-cell resolution, integrin subunit alpha X (ITGAX) was highly expressed in microglia, where specific subpopulations were associated with AD and cerebral amyloid angiopathy. DISCUSSION The study provides evidence of SPP1 and ITGAX association with cognitive decline and common neuropathologies identifying a microglial subset associated with disease.
Collapse
Affiliation(s)
- Katia de Paiva Lopes
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of Neurological SciencesRush University Medical CenterChicagoIllinoisUSA
| | - Lei Yu
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of Neurological SciencesRush University Medical CenterChicagoIllinoisUSA
| | - Xianli Shen
- Department of Cancer Immunology and VirologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
- Department of ImmunologyHarvard Medical SchoolBostonMassachusettsUSA
| | - Yiguo Qiu
- Department of Cancer Immunology and VirologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
- Department of ImmunologyHarvard Medical SchoolBostonMassachusettsUSA
- Chongqing International Institute for ImmunologyChongqingChina
| | - Shinya Tasaki
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of Neurological SciencesRush University Medical CenterChicagoIllinoisUSA
| | - Artemis Iatrou
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of Neurological SciencesRush University Medical CenterChicagoIllinoisUSA
- Department of Psychiatry, McLean HospitalHarvard Medical SchoolBelmontMassachusettsUSA
| | - Michal Schnaider Beeri
- Joseph Sagol Neuroscience Center, Sheba Medical CenterRamat GanIsrael
- Department of PsychiatryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- The Herbert and Jackeline Krieger Klein Alzheimer's Research CenterRutgers Biomedical and Health Sciences, Rutgers UniversityNew JerseyUSA
| | - Nicholas T. Seyfried
- Goizueta Alzheimer's Disease Research Center, Department of Neurology and Department of BiochemistryEmory University School of MedicineAtlantaGeorgiaUSA
| | - Vilas Menon
- Center for Translational and Computational NeuroimmunologyDepartment of Neurology & Taub Institute for Research on Alzheimer's disease and the Aging BrainColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Yanling Wang
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of Neurological SciencesRush University Medical CenterChicagoIllinoisUSA
| | - Julie A. Schneider
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of Neurological SciencesRush University Medical CenterChicagoIllinoisUSA
- Department of PathologyRush University Medical CenterChicagoIllinoisUSA
| | - Harvey Cantor
- Department of Cancer Immunology and VirologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
- Department of ImmunologyHarvard Medical SchoolBostonMassachusettsUSA
| | - David A. Bennett
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of Neurological SciencesRush University Medical CenterChicagoIllinoisUSA
| |
Collapse
|
14
|
Mendes EA, Tang Y, Jiang G. The integrated stress response signaling during the persistent HIV infection. iScience 2023; 26:108418. [PMID: 38058309 PMCID: PMC10696111 DOI: 10.1016/j.isci.2023.108418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2023] Open
Abstract
Human immunodeficiency virus-1 (HIV) infection is a chronic disease under antiretroviral therapy (ART), during which active HIV replication is effectively suppressed. Stable viral reservoirs are established early in infection and cannot be eradicated in people with HIV (PWH) by ART alone, which features residual immune inflammation with disease-associated secondary comorbidities. Mammalian cells are equipped with integrated stress response (ISR) machinery to detect intrinsic and extrinsic stresses such as heme deficiency, nutrient fluctuation, the accumulation of unfolded proteins, and viral infection. ISR is the part of the innate immunity that defends against pathogen infection or environmental alteration, thereby maintaining homeostasis to avoid diseases. Here, we describe how this machinery responds to the off-target effects of ART and persistent HIV infection in both the peripheral compartments and the brain. The latter may be important for us to better understand the mechanisms of stable HIV reservoirs and HIV-associated neurocognitive disorders.
Collapse
Affiliation(s)
- Erica A. Mendes
- UNC HIV Cure Center, Institute of Global Health and Infectious Diseases, the University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7042, USA
| | - Yuyang Tang
- UNC HIV Cure Center, Institute of Global Health and Infectious Diseases, the University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7042, USA
| | - Guochun Jiang
- UNC HIV Cure Center, Institute of Global Health and Infectious Diseases and the Department of Biochemistry and Biophysics, the University of North Carolina at Chapel Hill, Chapel Hill, NC 27599- 7042, USA
| |
Collapse
|
15
|
Banga R, Procopio FA, Lana E, Gladkov GT, Roseto I, Parsons EM, Lian X, Armani-Tourret M, Bellefroid M, Gao C, Kauzlaric A, Foglierini M, Alfageme-Abello O, Sluka SHM, Munoz O, Mastrangelo A, Fenwick C, Muller Y, Mkindi CG, Daubenberger C, Cavassini M, Trunfio R, Déglise S, Corpataux JM, Delorenzi M, Lichterfeld M, Pantaleo G, Perreau M. Lymph node dendritic cells harbor inducible replication-competent HIV despite years of suppressive ART. Cell Host Microbe 2023; 31:1714-1731.e9. [PMID: 37751747 PMCID: PMC11068440 DOI: 10.1016/j.chom.2023.08.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 06/02/2023] [Accepted: 08/30/2023] [Indexed: 09/28/2023]
Abstract
Although gut and lymph node (LN) memory CD4 T cells represent major HIV and simian immunodeficiency virus (SIV) tissue reservoirs, the study of the role of dendritic cells (DCs) in HIV persistence has long been limited to the blood due to difficulties to access lymphoid tissue samples. In this study, we show that LN migratory and resident DC subpopulations harbor distinct phenotypic and transcriptomic profiles. Interestingly, both LN DC subpopulations contain HIV intact provirus and inducible replication-competent HIV despite the expression of the antiviral restriction factor SAMHD1. Notably, LN DC subpopulations isolated from HIV-infected individuals treated for up to 14 years are transcriptionally silent but harbor replication-competent virus that can be induced upon TLR7/8 stimulation. Taken together, these results uncover a potential important contribution of LN DCs to HIV infection in the presence of ART.
Collapse
Affiliation(s)
- Riddhima Banga
- Services of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Francesco Andrea Procopio
- Services of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Erica Lana
- Services of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | | | | | - Elizabeth M Parsons
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA; Infectious Disease Division, Brigham and Women's Hospital, Boston, MA, USA
| | - Xiaodong Lian
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA; Infectious Disease Division, Brigham and Women's Hospital, Boston, MA, USA
| | | | | | - Ce Gao
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Annamaria Kauzlaric
- Translational Bioinformatics and Statistics Department of Oncology, University of Lausanne Swiss Cancer Center, Lausanne, Switzerland
| | - Mathilde Foglierini
- Services of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Oscar Alfageme-Abello
- Services of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Susanna H M Sluka
- Newborn Screening Switzerland, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Olivia Munoz
- Services of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Andrea Mastrangelo
- Services of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Craig Fenwick
- Services of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Yannick Muller
- Services of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Catherine Gerald Mkindi
- Ifakara Health Institute, Bagamoyo, United Republic of Tanzania; Department of Medical Parasitology and Infection Biology, Clinical Immunology Unit, Swiss Tropical and Public Health Institute, Basel, Switzerland
| | - Claudia Daubenberger
- Department of Medical Parasitology and Infection Biology, Clinical Immunology Unit, Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, 4001 Basel, Switzerland
| | - Matthias Cavassini
- Services of Infectious Diseases, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Rafael Trunfio
- Services of Vascular Surgery, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Sébastien Déglise
- Services of Vascular Surgery, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Jean-Marc Corpataux
- Services of Vascular Surgery, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Mauro Delorenzi
- Translational Bioinformatics and Statistics Department of Oncology, University of Lausanne Swiss Cancer Center, Lausanne, Switzerland
| | - Mathias Lichterfeld
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA; Infectious Disease Division, Brigham and Women's Hospital, Boston, MA, USA
| | - Giuseppe Pantaleo
- Services of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland; Swiss Vaccine Research Institute, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Matthieu Perreau
- Services of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland.
| |
Collapse
|
16
|
Downie LE, Zhang X, Wu M, Karunaratne S, Loi JK, Senthil K, Arshad S, Bertram K, Cunningham AL, Carnt N, Mueller SN, Chinnery HR. Redefining the human corneal immune compartment using dynamic intravital imaging. Proc Natl Acad Sci U S A 2023; 120:e2217795120. [PMID: 37487076 PMCID: PMC10400993 DOI: 10.1073/pnas.2217795120] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 06/13/2023] [Indexed: 07/26/2023] Open
Abstract
The healthy human cornea is a uniquely transparent sensory tissue where immune responses are tightly controlled to preserve vision. The cornea contains immune cells that are widely presumed to be intraepithelial dendritic cells (DCs). Corneal immune cells have diverse cellular morphologies and morphological alterations are used as a marker of inflammation and injury. Based on our imaging of corneal T cells in mice, we hypothesized that many human corneal immune cells commonly defined as DCs are intraepithelial lymphocytes (IELs). To investigate this, we developed functional in vivo confocal microscopy (Fun-IVCM) to investigate cell dynamics in the human corneal epithelium and stroma. We show that many immune cells resident in the healthy human cornea are T cells. These corneal IELs are characterized by rapid, persistent motility and interact with corneal DCs and sensory nerves. Imaging deeper into the corneal stroma, we show that crawling macrophages and rare motile T cells patrol the tissue. Furthermore, we identify altered immune cell behaviors in response to short-term contact lens wear (acute inflammatory stimulus), as well as in individuals with allergy (chronic inflammatory stimulus) that was modulated by therapeutic intervention. These findings redefine current understanding of immune cell subsets in the human cornea and reveal how resident corneal immune cells respond and adapt to chronic and acute stimuli.
Collapse
Affiliation(s)
- Laura E. Downie
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, VIC3053, Australia
| | - Xinyuan Zhang
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, VIC3053, Australia
| | - Mengliang Wu
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, VIC3053, Australia
| | - Senuri Karunaratne
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, VIC3053, Australia
| | - Joon Keit Loi
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC3010, Australia
| | - Kirthana Senthil
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, VIC3053, Australia
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC3010, Australia
| | - Sana Arshad
- The Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW2145, Australia
| | - Kirstie Bertram
- The Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW2145, Australia
| | - Anthony L. Cunningham
- The Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW2145, Australia
| | - Nicole Carnt
- The Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW2145, Australia
- School of Optometry and Vision Science, University of New South Wales, Kensington, NSW2052, Australia
- Institute of Ophthalmology, University College London, LondonEC1V 9EL, United Kingdom
| | - Scott N. Mueller
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC3010, Australia
| | - Holly R. Chinnery
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, VIC3053, Australia
| |
Collapse
|
17
|
Lang M, Krump C, Meshcheryakova A, Tam-Amersdorfer C, Schwarzenberger E, Passegger C, Connolly S, Mechtcheriakova D, Strobl H. Microenvironmental and cell intrinsic factors governing human cDC2 differentiation and monocyte reprogramming. Front Immunol 2023; 14:1216352. [PMID: 37539048 PMCID: PMC10395083 DOI: 10.3389/fimmu.2023.1216352] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 06/14/2023] [Indexed: 08/05/2023] Open
Abstract
cDC2s occur abundantly in peripheral tissues and arise from circulating blood cDC2s. However, the factors governing cDC2 differentiation in tissues, especially under inflammatory conditions, remained poorly defined. We here found that psoriatic cDC2s express the efferocytosis receptor Axl and exhibit a bone morphogenetic protein (BMP) and p38MAPK signaling signature. BMP7, strongly expressed within the lesional psoriatic epidermis, cooperates with canonical TGF-β1 signaling for inducing Axl+cDC2s from blood cDC2s in vitro. Moreover, downstream induced p38MAPK promotes Axl+cDC2s at the expense of Axl+CD207+ Langerhans cell differentiation from blood cDC2s. BMP7 supplementation allowed to model cDC2 generation and their further differentiation into LCs from CD34+ hematopoietic progenitor cells in defined serum-free medium. Additionally, p38MAPK promoted the generation of another cDC2 subset lacking Axl but expressing the non-classical NFkB transcription factor RelB in vitro. Such RelB+cDC2s occurred predominantly at dermal sites in the inflamed skin. Finally, we found that cDC2s can be induced to acquire high levels of the monocyte lineage identity factor kruppel-like-factor-4 (KLF4) along with monocyte-derived DC and macrophage phenotypic characteristics in vitro. In conclusion, inflammatory and psoriatic epidermal signals instruct blood cDC2s to acquire phenotypic characteristics of several tissue-resident cell subsets.
Collapse
Affiliation(s)
- Magdalena Lang
- Division of Immunology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Corinna Krump
- Division of Immunology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Anastasia Meshcheryakova
- Insitute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Carmen Tam-Amersdorfer
- Division of Immunology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Elke Schwarzenberger
- Division of Immunology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Christina Passegger
- Division of Immunology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Sally Connolly
- Division of Immunology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Diana Mechtcheriakova
- Insitute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Herbert Strobl
- Division of Immunology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| |
Collapse
|
18
|
Mariotton J, Cohen E, Zhu A, Auffray C, Barbosa Bomfim CC, Barry Delongchamps N, Zerbib M, Bomsel M, Ganor Y. TRPV1 activation in human Langerhans cells and T cells inhibits mucosal HIV-1 infection via CGRP-dependent and independent mechanisms. Proc Natl Acad Sci U S A 2023; 120:e2302509120. [PMID: 37216549 PMCID: PMC10235960 DOI: 10.1073/pnas.2302509120] [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: 02/28/2023] [Accepted: 04/27/2023] [Indexed: 05/24/2023] Open
Abstract
Upon its mucosal transmission, HIV type 1 (HIV-1) rapidly targets genital antigen-presenting Langerhans cells (LCs), which subsequently transfer infectious virus to CD4+ T cells. We previously described an inhibitory neuroimmune cross talk, whereby calcitonin gene-related peptide (CGRP), a neuropeptide secreted by peripheral pain-sensing nociceptor neurons innervating all mucosal epithelia and associating with LCs, strongly inhibits HIV-1 transfer. As nociceptors secret CGRP following the activation of their Ca2+ ion channel transient receptor potential vanilloid 1 (TRPV1), and as we reported that LCs secret low levels of CGRP, we investigated whether LCs express functional TRPV1. We found that human LCs expressed mRNA and protein of TRPV1, which was functional and induced Ca2+ influx following activation with TRPV1 agonists, including capsaicin (CP). The treatment of LCs with TRPV1 agonists also increased CGRP secretion, reaching its anti-HIV-1 inhibitory concentrations. Accordingly, CP pretreatment significantly inhibited LCs-mediated HIV-1 transfer to CD4+ T cells, which was abrogated by both TRPV1 and CGRP receptor antagonists. Like CGRP, CP-induced inhibition of HIV-1 transfer was mediated via increased CCL3 secretion and HIV-1 degradation. CP also inhibited direct CD4+ T cells HIV-1 infection, but in CGRP-independent manners. Finally, pretreatment of inner foreskin tissue explants with CP markedly increased CGRP and CCL3 secretion, and upon subsequent polarized exposure to HIV-1, inhibited an increase in LC-T cell conjugate formation and consequently T cell infection. Our results reveal that TRPV1 activation in human LCs and CD4+ T cells inhibits mucosal HIV-1 infection, via CGRP-dependent/independent mechanisms. Formulations containing TRPV1 agonists, already approved for pain relief, could hence be useful against HIV-1.
Collapse
Affiliation(s)
- Jammy Mariotton
- Laboratory of Mucosal Entry of HIV-1 and Mucosal Immunity, Department of infection Immunity and Inflammation, Universiteé Paris Cité, Institut Cochin, INSERM U1016, CNRS UMR8104, F-75014Paris, France
| | - Emmanuel Cohen
- Laboratory of Mucosal Entry of HIV-1 and Mucosal Immunity, Department of infection Immunity and Inflammation, Universiteé Paris Cité, Institut Cochin, INSERM U1016, CNRS UMR8104, F-75014Paris, France
| | - Aiwei Zhu
- Laboratory of Mucosal Entry of HIV-1 and Mucosal Immunity, Department of infection Immunity and Inflammation, Universiteé Paris Cité, Institut Cochin, INSERM U1016, CNRS UMR8104, F-75014Paris, France
| | - Cédric Auffray
- Laboratory of Regulation of T Cell Effector Functions, Department of infection Immunity and Inflammation, Universiteé Paris Cité, Institut Cochin, INSERM U1016, CNRS UMR8104, F-75014Paris, France
| | - Caio César Barbosa Bomfim
- Laboratory of Mucosal Entry of HIV-1 and Mucosal Immunity, Department of infection Immunity and Inflammation, Universiteé Paris Cité, Institut Cochin, INSERM U1016, CNRS UMR8104, F-75014Paris, France
| | | | - Marc Zerbib
- Urology Service, Groupe Hospitalier (GH) Cochin-St Vincent de Paul, F-75014Paris, France
| | - Morgane Bomsel
- Laboratory of Mucosal Entry of HIV-1 and Mucosal Immunity, Department of infection Immunity and Inflammation, Universiteé Paris Cité, Institut Cochin, INSERM U1016, CNRS UMR8104, F-75014Paris, France
| | - Yonatan Ganor
- Laboratory of Mucosal Entry of HIV-1 and Mucosal Immunity, Department of infection Immunity and Inflammation, Universiteé Paris Cité, Institut Cochin, INSERM U1016, CNRS UMR8104, F-75014Paris, France
| |
Collapse
|
19
|
Wu Y, Li M, Zhang J, Wang S. Unveiling uterine aging: Much more to learn. Ageing Res Rev 2023; 86:101879. [PMID: 36764360 DOI: 10.1016/j.arr.2023.101879] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/22/2023] [Accepted: 02/05/2023] [Indexed: 02/11/2023]
Abstract
Uterine aging is an important factor that impacts fertility, reproductive health, and uterus-related diseases; however, it remains poorly explored. Functionally, these disturbances have been associated with an abnormal hormonal response in the endometrium and decreased endometrial receptivity. Based on emerging evidence, these alterations are mediated via the senescence of endometrial stem cells and impaired decidualization of endometrial stromal cells. Multiple molecular activities may participate in uterine aging, including oxidative stress, inflammation, fibrosis, DNA damage response, and cellular senescence. Over the past decade, several protective strategies targeting these biological processes have afforded promising results, including stem cell therapy, anti-aging drugs, and herbal medicines. However, the currently available evidence is fragmented and scattered. Here, we summarize the most recent findings regarding uterine aging, including functional and structural alterations and potential cellular and molecular mechanisms, and discuss potential protective interventions against uterine aging. Thereby, we hope to provide a comprehensive understanding of the pathophysiological processes and underlying mechanisms associated with uterine aging, as well as improve fecundity and reproductive outcomes in females of advanced reproductive age.
Collapse
Affiliation(s)
- Yaling Wu
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Milu Li
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinjin Zhang
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Shixuan Wang
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
20
|
Zhu R, Liu X, Li X, Yao X, Li W. Response to identifying the epidermal dendritic cell landscape. Immunity 2023; 56:461-462. [PMID: 36921568 DOI: 10.1016/j.immuni.2023.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Affiliation(s)
- Ronghui Zhu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Xiaochun Liu
- Department of Allergy and Rheumatology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing 210042, P.R. China
| | - Xiao Li
- Gene Editing Laboratory, Texas Heart Institute, Houston, TX 77030, USA
| | - Xu Yao
- Department of Allergy and Rheumatology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing 210042, P.R. China.
| | - Wei Li
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China.
| |
Collapse
|
21
|
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
|
22
|
van Teijlingen NH, Eder J, Sarrami-Forooshani R, Zijlstra-Willems EM, Roovers JPWR, van Leeuwen E, Ribeiro CMS, Geijtenbeek TBH. Immune activation of vaginal human Langerhans cells increases susceptibility to HIV-1 infection. Sci Rep 2023; 13:3283. [PMID: 36841916 PMCID: PMC9968315 DOI: 10.1038/s41598-023-30097-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 02/14/2023] [Indexed: 02/27/2023] Open
Abstract
Vaginal inflammation increases the risk for sexual HIV-1 transmission but underlying mechanisms remain unclear. In this study we assessed the impact of immune activation on HIV-1 susceptibility of primary human vaginal Langerhans cells (LCs). Vaginal LCs isolated from human vaginal tissue expressed a broad range of TLRs and became activated after exposure to both viral and bacterial TLR ligands. HIV-1 replication was restricted in immature vaginal LCs as only low levels of infection could be detected. Notably, activation of immature vaginal LCs by bacterial TLR ligands increased HIV-1 infection, whereas viral TLR ligands were unable to induce HIV-1 replication in vaginal LCs. Furthermore, mature vaginal LCs transmitted HIV-1 to CD4 T cells. This study emphasizes the role for vaginal LCs in protection against mucosal HIV-1 infection, which is abrogated upon activation. Moreover, our data suggest that bacterial STIs can increase the risk of HIV-1 acquisition in women.
Collapse
Affiliation(s)
- Nienke H. van Teijlingen
- grid.509540.d0000 0004 6880 3010Amsterdam UMC Location Academic Medical Center, Experimental Immunology, Meibergdreef 9, Amsterdam, The Netherlands
| | - Julia Eder
- grid.509540.d0000 0004 6880 3010Amsterdam UMC Location Academic Medical Center, Experimental Immunology, Meibergdreef 9, Amsterdam, The Netherlands ,Amsterdam Institute for Infection & Immunity, Amsterdam, The Netherlands
| | - Ramin Sarrami-Forooshani
- grid.417689.5ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, P.O. BOX, Tehran, 15179/64311 Iran
| | - Esther M. Zijlstra-Willems
- grid.509540.d0000 0004 6880 3010Amsterdam UMC Location Academic Medical Center, Experimental Immunology, Meibergdreef 9, Amsterdam, The Netherlands ,Amsterdam Institute for Infection & Immunity, Amsterdam, The Netherlands
| | - Jan-Paul W. R. Roovers
- grid.509540.d0000 0004 6880 3010Amsterdam UMC Location Academic Medical Center, Obstetrics and Gynaecology, Meibergdreef 9, Amsterdam, The Netherlands
| | - Elisabeth van Leeuwen
- grid.509540.d0000 0004 6880 3010Amsterdam UMC Location Academic Medical Center, Obstetrics and Gynaecology, Meibergdreef 9, Amsterdam, The Netherlands
| | - Carla M. S. Ribeiro
- grid.509540.d0000 0004 6880 3010Amsterdam UMC Location Academic Medical Center, Experimental Immunology, Meibergdreef 9, Amsterdam, The Netherlands ,Amsterdam Institute for Infection & Immunity, Amsterdam, The Netherlands
| | - Teunis B. H. Geijtenbeek
- grid.509540.d0000 0004 6880 3010Amsterdam UMC Location Academic Medical Center, Experimental Immunology, Meibergdreef 9, Amsterdam, The Netherlands ,Amsterdam Institute for Infection & Immunity, Amsterdam, The Netherlands
| |
Collapse
|
23
|
Andre M, Nair M, Raymond AD. HIV Latency and Nanomedicine Strategies for Anti-HIV Treatment and Eradication. Biomedicines 2023; 11:biomedicines11020617. [PMID: 36831153 PMCID: PMC9953021 DOI: 10.3390/biomedicines11020617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/09/2023] [Accepted: 02/12/2023] [Indexed: 02/22/2023] Open
Abstract
Antiretrovirals (ARVs) reduce Human Immunodeficiency Virus (HIV) loads to undetectable levels in infected patients. However, HIV can persist throughout the body in cellular reservoirs partly due to the inability of some ARVs to cross anatomical barriers and the capacity of HIV-1 to establish latent infection in resting CD4+ T cells and monocytes/macrophages. A cure for HIV is not likely unless latency is addressed and delivery of ARVs to cellular reservoir sites is improved. Nanomedicine has been used in ARV formulations to improve delivery and efficacy. More specifically, researchers are exploring the benefit of using nanoparticles to improve ARVs and nanomedicine in HIV eradication strategies such as shock and kill, block and lock, and others. This review will focus on mechanisms of HIV-1 latency and nanomedicine-based approaches to treat HIV.
Collapse
Affiliation(s)
- Mickensone Andre
- Department of Immunology and Nanomedicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Madhavan Nair
- Department of Immunology and Nanomedicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
- Institute of Neuroimmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Andrea D. Raymond
- Department of Immunology and Nanomedicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
- Correspondence: ; Tel.: +1-305-348-6430
| |
Collapse
|
24
|
Poondla N, Sheykhhasan M, Ahmadyousefi Y, Akbari M, Seyedebrahimi R, Farsani ME, Kalhor N. Dendritic Cells - Winning the Fight against HIV. Curr Stem Cell Res Ther 2023; 18:174-185. [PMID: 35366782 DOI: 10.2174/1574888x17666220401102718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/11/2022] [Accepted: 02/09/2022] [Indexed: 11/22/2022]
Abstract
HIV is a virus that targets and hijacks the immune cells of the host. It multiplies by attacking the helper T-lymphocytes. HIV has remained one of the most difficult and dangerous infections in the world due to the inability to find a successful treatment and a lack of access to medical care. When the virus reaches the body, dendritic cells are the first cells it encounters. DCs have been identified as one of the most effective mediators of immune responses, implying a promising strategy against viral infection. The current state of knowledge about the function of dendritic cells and their subsets is critical for using their full potential as a candidate for the development of an HIV vaccine. Despite extensive efforts, a reliable vaccine with the fewest side effects has yet to be found, and further research is needed to find a dependable and efficient vaccine. The extent to which dendritic cell-based therapy is used to treat HIV was investigated in this study. As the virus attacks the host immune system, the dendritic cells can trigger an immune response against HIV-1 infection.
Collapse
Affiliation(s)
- Naresh Poondla
- Icahn School of Medicine at Mount Sinai, New York, United States
| | - Mohsen Sheykhhasan
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture and Research [ACECR], Qom Branch, Qom, Iran
| | - Yaghoub Ahmadyousefi
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Akbari
- Department of Medical School, Faculty of Medical Sciences, Islamic Azad University, Tonekabon Branch, Mazandaran, Iran
| | | | - Mohsen Eslami Farsani
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
| | - Naser Kalhor
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture and Research [ACECR], Qom Branch, Qom, Iran
| |
Collapse
|
25
|
Karim QA, Archary D, Barré-Sinoussi F, Broliden K, Cabrera C, Chiodi F, Fidler SJ, Gengiah TN, Herrera C, Kharsany ABM, Liebenberg LJP, Mahomed S, Menu E, Moog C, Scarlatti G, Seddiki N, Sivro A, Cavarelli M. Women for science and science for women: Gaps, challenges and opportunities towards optimizing pre-exposure prophylaxis for HIV-1 prevention. Front Immunol 2022; 13:1055042. [PMID: 36561760 PMCID: PMC9763292 DOI: 10.3389/fimmu.2022.1055042] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 11/11/2022] [Indexed: 12/12/2022] Open
Abstract
Preventing new HIV infections remains a global challenge. Young women continue to bear a disproportionate burden of infection. Oral pre-exposure prophylaxis (PrEP), offers a novel women-initiated prevention technology and PrEP trials completed to date underscore the importance of their inclusion early in trials evaluating new HIV PrEP technologies. Data from completed topical and systemic PrEP trials highlight the role of gender specific physiological and social factors that impact PrEP uptake, adherence and efficacy. Here we review the past and current developments of HIV-1 prevention options for women with special focus on PrEP considering the diverse factors that can impact PrEP efficacy. Furthermore, we highlight the importance of inclusion of female scientists, clinicians, and community advocates in scientific efforts to further improve HIV prevention strategies.
Collapse
Affiliation(s)
- Quarraisha Abdool Karim
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Doris Duke Medical Research Institute (2Floor), Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, United States
| | - Derseree Archary
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Doris Duke Medical Research Institute (2Floor), Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
- Department of Medical Microbiology, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | | | - Kristina Broliden
- Department of Medicine Solna, Division of Infectious Diseases, Karolinska Institutet, Department of Infectious Diseases, Karolinska University Hospital, Center for Molecular Medicine, Stockholm, Sweden
| | - Cecilia Cabrera
- AIDS Research Institute IrsiCaixa, Institut de Recerca en Ciències de la Salut Germans Trias i Pujol (IGTP), Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Francesca Chiodi
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Sarah J. Fidler
- Department of Infectious Disease, Faculty of Medicine, Imperial College London UK and Imperial College NIHR BRC, London, United Kingdom
| | - Tanuja N. Gengiah
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Doris Duke Medical Research Institute (2Floor), Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Carolina Herrera
- Department of Infectious Disease, Section of Virology, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Ayesha B. M. Kharsany
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Doris Duke Medical Research Institute (2Floor), Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
- Department of Medical Microbiology, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Lenine J. P. Liebenberg
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Doris Duke Medical Research Institute (2Floor), Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
- Department of Medical Microbiology, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Sharana Mahomed
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Doris Duke Medical Research Institute (2Floor), Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Elisabeth Menu
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
- MISTIC Group, Department of Virology, Institut Pasteur, Paris, France
| | - Christiane Moog
- Laboratoire d’ImmunoRhumatologie Moléculaire, Institut national de la santé et de la recherche médicale (INSERM) UMR_S 1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantex NG, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Gabriella Scarlatti
- Viral Evolution and Transmission Unit, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Nabila Seddiki
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Aida Sivro
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Doris Duke Medical Research Institute (2Floor), Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
- Department of Medical Microbiology, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
- JC Wilt Infectious Disease Research Centre, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Mariangela Cavarelli
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| |
Collapse
|
26
|
Smith JB, Herbert JJ, Truong NR, Cunningham AL. Cytokines and chemokines: The vital role they play in herpes simplex virus mucosal immunology. Front Immunol 2022; 13:936235. [PMID: 36211447 PMCID: PMC9538770 DOI: 10.3389/fimmu.2022.936235] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 09/08/2022] [Indexed: 11/17/2022] Open
Abstract
Herpes simplex viruses (HSV) types 1 and 2 are ubiquitous infections in humans. They cause orofacial and genital herpes with occasional severe complications. HSV2 also predisposes individuals to infection with HIV. There is currently no vaccine or immunotherapy for these diseases. Understanding the immunopathogenesis of HSV infections is essential to progress towards these goals. Both HSV viruses result in initial infections in two major sites - in the skin or mucosa, either after initial infection or recurrence, and in the dorsal root or trigeminal ganglia where the viruses establish latency. HSV1 can also cause recurrent infection in the eye. At all of these sites immune cells respond to control infection. T cells and resident dendritic cells (DCs) in the skin/mucosa and around reactivating neurones in the ganglia, as well as keratinocytes in the skin and mucosa, are major sources of cytokines and chemokines. Cytokines such as the Type I and II interferons synergise in their local antiviral effects. Chemokines such as CCL2, 3 and 4 are found in lesion vesicle fluid, but their exact role in determining the interactions between epidermal and dermal DCs and with resident memory and infiltrating CD4 and CD8 T cells in the skin/mucosa is unclear. Even less is known about these mechanisms in the ganglia. Here we review the data on known sources and actions of these cytokines and chemokines at cellular and tissue level and indicate their potential for preventative and therapeutic interventions.
Collapse
Affiliation(s)
- Jacinta B. Smith
- Centre for Virus Research, The Westmead Institute for Medical Research, Sydney, NSW, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Jason J. Herbert
- Centre for Virus Research, The Westmead Institute for Medical Research, Sydney, NSW, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Naomi R. Truong
- Centre for Virus Research, The Westmead Institute for Medical Research, Sydney, NSW, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Anthony L. Cunningham
- Centre for Virus Research, The Westmead Institute for Medical Research, Sydney, NSW, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- *Correspondence: Anthony L. Cunningham,
| |
Collapse
|
27
|
Baharlou H, Canete N, Vine EE, Hu K, Yuan D, Sandgren KJ, Bertram KM, Nasr N, Rhodes JW, Gosselink MP, Di Re A, Reza F, Ctercteko G, Pathma-Nathan N, Collins G, Toh J, Patrick E, Haniffa MA, Estes JD, Byrne SN, Cunningham AL, Harman AN. An in situ analysis pipeline for initial host-pathogen interactions reveals signatures of human colorectal HIV transmission. Cell Rep 2022; 40:111385. [PMID: 36130503 DOI: 10.1016/j.celrep.2022.111385] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 07/07/2022] [Accepted: 08/29/2022] [Indexed: 12/01/2022] Open
Abstract
The initial immune response to HIV determines transmission. However, due to technical limitations we still do not have a comparative map of early mucosal transmission events. By combining RNAscope, cyclic immunofluorescence, and image analysis tools, we quantify HIV transmission signatures in intact human colorectal explants within 2 h of topical exposure. We map HIV enrichment to mucosal dendritic cells (DCs) and submucosal macrophages, but not CD4+ T cells, the primary targets of downstream infection. HIV+ DCs accumulate near and within lymphoid aggregates, which act as early sanctuaries of high viral titers while facilitating HIV passage to the submucosa. Finally, HIV entry induces recruitment and clustering of target cells, facilitating DC- and macrophage-mediated HIV transfer and enhanced infection of CD4+ T cells. These data demonstrate a rapid response to HIV structured to maximize the likelihood of mucosal infection and provide a framework for in situ studies of host-pathogen interactions and immune-mediated pathologies.
Collapse
Affiliation(s)
- Heeva Baharlou
- Centre for Virus Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW 2145, Australia; The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health Sydney, Sydney, NSW, Australia.
| | - Nicolas Canete
- Centre for Virus Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW 2145, Australia; The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health Sydney, Sydney, NSW, Australia
| | - Erica E Vine
- Centre for Virus Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW 2145, Australia; The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health Sydney, Sydney, NSW, Australia
| | - Kevin Hu
- Centre for Virus Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW 2145, Australia; The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health Sydney, Sydney, NSW, Australia
| | - Di Yuan
- Centre for Virus Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW 2145, Australia; The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health Sydney, Sydney, NSW, Australia
| | - Kerrie J Sandgren
- Centre for Virus Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW 2145, Australia; The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health Sydney, Sydney, NSW, Australia
| | - Kirstie M Bertram
- Centre for Virus Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW 2145, Australia; The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health Sydney, Sydney, NSW, Australia
| | - Najla Nasr
- Centre for Virus Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW 2145, Australia; The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health Sydney, Sydney, NSW, Australia
| | - Jake W Rhodes
- Centre for Virus Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW 2145, Australia; The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health Sydney, Sydney, NSW, Australia
| | - Martijn P Gosselink
- Centre for Virus Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW 2145, Australia; Department of Colorectal Surgery, Westmead Hospital, Westmead, NSW 2145, Australia
| | - Angelina Di Re
- Centre for Virus Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW 2145, Australia; Department of Colorectal Surgery, Westmead Hospital, Westmead, NSW 2145, Australia
| | - Faizur Reza
- Centre for Virus Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW 2145, Australia; Department of Colorectal Surgery, Westmead Hospital, Westmead, NSW 2145, Australia
| | - Grahame Ctercteko
- Centre for Virus Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW 2145, Australia; Department of Colorectal Surgery, Westmead Hospital, Westmead, NSW 2145, Australia
| | - Nimalan Pathma-Nathan
- Centre for Virus Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW 2145, Australia; Department of Colorectal Surgery, Westmead Hospital, Westmead, NSW 2145, Australia
| | - Geoff Collins
- Centre for Virus Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW 2145, Australia; Department of Colorectal Surgery, Westmead Hospital, Westmead, NSW 2145, Australia
| | - James Toh
- Centre for Virus Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW 2145, Australia; Department of Colorectal Surgery, Westmead Hospital, Westmead, NSW 2145, Australia
| | - Ellis Patrick
- Centre for Virus Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW 2145, Australia; The University of Sydney, School of Maths and Statistics, Faculty of Science, Sydney, 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
| | - Jacob D Estes
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Portland, OR, USA; Division of Pathobiology & Immunology, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Scott N Byrne
- Centre for Virus Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW 2145, Australia; The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health Sydney, Sydney, NSW, Australia
| | - Anthony L Cunningham
- Centre for Virus Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW 2145, Australia; The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health Sydney, Sydney, NSW, Australia
| | - Andrew N Harman
- Centre for Virus Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW 2145, Australia; The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health Sydney, Sydney, NSW, Australia.
| |
Collapse
|
28
|
Xuan S, Li Y, Wu Y, Adcock IM, Zeng X, Yao X. Langerin-expressing dendritic cells in pulmonary immune-related diseases. Front Med (Lausanne) 2022; 9:909057. [PMID: 36160158 PMCID: PMC9490018 DOI: 10.3389/fmed.2022.909057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 08/12/2022] [Indexed: 11/30/2022] Open
Abstract
Dendritic cells (DCs) are “frontline” immune cells dedicated to antigen presentation. They serve as an important bridge connecting innate and adaptive immunity, and express various receptors for antigen capture. DCs are divided into various subclasses according to their differential expression of cell surface receptors and different subclasses of DCs exhibit specific immunological characteristics. Exploring the common features of each sub-category has became the focus of many studies. There are certain amounts of DCs expressing langerin in airways and peripheral lungs while the precise mechanism by which langerin+ DCs drive pulmonary disease is unclear. Langerin-expressing DCs can be further subdivided into numerous subtypes based on the co-expressed receptors, but here, we identify commonalities across these subtypes that point to the major role of langerin. Better understanding is required to clarify key disease pathways and determine potential new therapeutic approaches.
Collapse
Affiliation(s)
- Shurui Xuan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuebei Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yunhui Wu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ian M. Adcock
- Airway Disease Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Xiaoning Zeng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xin Yao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Xin Yao
| |
Collapse
|
29
|
Saito M, Rajesh A, Innes C, van der Griend R, Fitzgerald P, Simcock B, Sykes P, Hibma M. Blimp-1 is a prognostic indicator for progression of cervical intraepithelial neoplasia grade 2. J Cancer Res Clin Oncol 2022; 148:1991-2002. [PMID: 35386001 PMCID: PMC9294030 DOI: 10.1007/s00432-022-03993-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 03/22/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND Progression of cervical intraepithelial neoplasia (CIN) to higher grade disease is associated with persistent human papillomavirus (HPV) infection and an absence of immune-mediated regression. However, the immune microenvironment that distinguishes progression from persistent or regressing lesions has not been well defined. METHODS A total of 69 patients under the age of 25 with high-risk HPV-positive cytology and biopsy-confirmed p16-positive CIN2 were included in the study. Biopsies were stained using 20 antibodies to a range of immune markers. Based on a 2-year follow-up, samples were analysed in "progressor" (CIN3 +) or "persister/regressor" (CIN1, 2 or normal) groups. RESULTS Progression was most strongly associated with Blimp-1 positive cell staining in the lesion (P = 0.0019) and with low numbers of infiltrating CD4 cells in the dermal region beneath the lesion (P = 0.0022). The presence of CD4, CD8 and T bet-positive cells in the dermal region most strongly correlated with CD11c cells in the persister/regressor but not the progressor group. CONCLUSION High numbers of Blimp-1 + cells in CIN2 lesions may predict progression to more severe disease. Measurement of Blimp-1 may have diagnostic utility for the determination of the need to treat women with cervical pre-cancer. HIGHLIGHTS CIN2 progression is associated with high numbers of Blimp-1 positive cells in the lesion. Detection of Blimp-1 in the lesion may have utility as a prognostic test to inform the need to treat CIN2.
Collapse
Affiliation(s)
- Mayumi Saito
- Department of Pathology, Dunedin School of Medicine, University of Otago, P O Box 56, Dunedin, New Zealand
| | - Aarthi Rajesh
- Department of Pathology, Dunedin School of Medicine, University of Otago, P O Box 56, Dunedin, New Zealand
| | - Carrie Innes
- Department of Obstetrics and Gynaecology, University of Otago, Christchurch, New Zealand
| | | | | | - Bryony Simcock
- Department of Obstetrics and Gynaecology, University of Otago, Christchurch, New Zealand
| | - Peter Sykes
- Department of Obstetrics and Gynaecology, University of Otago, Christchurch, New Zealand
| | - Merilyn Hibma
- Department of Pathology, Dunedin School of Medicine, University of Otago, P O Box 56, Dunedin, New Zealand.
| |
Collapse
|
30
|
Rajesh A, Saito M, Morrin H, Tschirley A, Simcock J, Currie M, Hibma M. Characterisation of the immune microenvironment of cutaneous squamous cell carcinoma in immunosuppression. Exp Dermatol 2022; 31:1720-1728. [PMID: 35861124 DOI: 10.1111/exd.14650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 06/23/2022] [Accepted: 07/18/2022] [Indexed: 11/29/2022]
Abstract
Cutaneous squamous cell carcinoma (cSCC) is a common cancer. Systemic immunosuppression with drugs such as Prednisone results in more aggressive disease. We hypothesise that more aggressive disease in immunosuppression is the result of immune changes in the tumour microenvironment. We characterised T cell, phagocytic and antigen presenting cell subsets in cSCC, and determined if these infiltrates were altered by immunosuppressive therapy. We found a dominant 'CD8 profile' in the centre of cSCC lesions, with CD8 cells correlating with Tbet, FoxP3, OX40 and 'M2-like' macrophages, whereas a 'Tbet and granulocyte profile' with associated inflammation predominated at the margin of the tumour. Individuals on systemic immunosuppressive therapy had lesions that were comparable in size, stage and number of vessels to immune competent individuals however the number of CD11c positive cells in the lesion centre was significantly reduced. We conclude that cSCC lesions are immunologically heterogeneous across the lesion and that systemically immunosuppressed individuals have reduced CD11c positive cells in the centre of the lesion. The role and detailed phenotype of CD11c positive cells in cSCC lesions warrants further investigation.
Collapse
Affiliation(s)
- Aarthi Rajesh
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Mayumi Saito
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Helen Morrin
- Cancer Society Tissue Bank, Christchurch, New Zealand
| | - Allison Tschirley
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Jeremy Simcock
- Department of Surgery, University of Otago Christchurch, Christchurch, New Zealand
| | - Margaret Currie
- Department of Pathology and Biomedical Science, University of Otago Christchurch, Christchurch, New Zealand
| | - Merilyn Hibma
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| |
Collapse
|
31
|
Mohammadi A, Bagherichimeh S, Choi Y, Fazel A, Tevlin E, Huibner S, Good SV, Tharao W, Kaul R. Immune parameters of HIV susceptibility in the female genital tract before and after penile-vaginal sex. COMMUNICATIONS MEDICINE 2022; 2:60. [PMID: 35637661 PMCID: PMC9142516 DOI: 10.1038/s43856-022-00122-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 05/03/2022] [Indexed: 11/09/2022] Open
Abstract
Background In women, most HIV infections are acquired through penile-vaginal sex. Inflammation in the female genital tract (FGT) increases the risk of HIV acquisition and transmission, likely through recruitment of HIV target cells and disruption of epithelial barrier integrity. Although sex may have important immune and epithelial effects, the impact of receptive penile-vaginal sex on the immune correlates of HIV susceptibility in the female genital tract is not well described. Methods STI-free heterosexual couples were recruited to the Sex, Couples and Science (SECS) Study, with the serial collection of cervical secretions (CVS), endocervical cytobrushes, blood and semen before and up to 72 h after either condomless (n = 29) or condom-protected (n = 8) penile-vaginal sex. Immune cells were characterized by flow cytometry, and immune factors including cytokines and soluble E-cadherin (sE-cad; a marker of epithelial disruption) were quantified by multiplex immunoassay. Co-primary endpoints were defined as levels of IP-10 and IL-1α, cytokines previously associated with increased HIV susceptibility. Results Here we show that cervicovaginal levels of vaginal IP-10, sE-cad and several other cytokines increase rapidly after sex, regardless of condom use. The proportion of endocervical HIV target cells, including Th17 cells, activated T cells, and activated or mature dendritic cells (DCs) also increase, particularly after condomless sex. Although most of these immune changes resolve within 72 h, increases in activated cervical CD4 + T cells and Tcm persist beyond this time. Conclusions Penile-vaginal sex induces multiple genital immune changes that may enhance HIV susceptibility during the 72 h post-sex window that is critical for virus acquisition. This has important implications for the mucosal immunopathogenesis of HIV transmission. Women who acquire HIV most commonly do so during penile-vaginal sex. Although the risk of HIV acquisition is higher when there is pre-existing inflammation in the female genital tract, the impact of receptive penile-vaginal sex itself on immune markers of HIV susceptibility in the genital tract has not been widely studied. We recruited heterosexual couples, without HIV or sexually-transmitted infections, and studied the impact of a single episode of penile-vaginal sex on immune cells and proteins in the female genital tract. We found that some markers within the cervix and vagina increased immediately after sex, then returned to normal. We noticed differences in these changes depending on whether the sex was condom-protected and whether the male partner was circumcised. Our findings might help us to understand how sex impacts the immune system and how this might contribute to HIV acquisition. Mohammadi et al. evaluate immune markers and cell types associated with HIV susceptibility in the female genital tract before and after penile-vaginal sex. The authors report that these immune parameters increase rapidly and transiently after sex, with condom use affecting some of the changes observed.
Collapse
|
32
|
York J, Gowrishankar K, Micklethwaite K, Palmer S, Cunningham AL, Nasr N. Evolving Strategies to Eliminate the CD4 T Cells HIV Viral Reservoir via CAR T Cell Immunotherapy. Front Immunol 2022; 13:873701. [PMID: 35572509 PMCID: PMC9098815 DOI: 10.3389/fimmu.2022.873701] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
Although the advent of ART has significantly reduced the morbidity and mortality associated with HIV infection, the stable pool of HIV in latently infected cells requires lifelong treatment adherence, with the cessation of ART resulting in rapid reactivation of the virus and productive HIV infection. Therefore, these few cells containing replication-competent HIV, known as the latent HIV reservoir, act as the main barrier to immune clearance and HIV cure. While several strategies involving HIV silencing or its reactivation in latently infected cells for elimination by immune responses have been explored, exciting cell based immune therapies involving genetically engineered T cells expressing synthetic chimeric receptors (CAR T cells) are highly appealing and promising. CAR T cells, in contrast to endogenous cytotoxic T cells, can function independently of MHC to target HIV-infected cells, are efficacious and have demonstrated acceptable safety profiles and long-term persistence in peripheral blood. In this review, we present a comprehensive picture of the current efforts to target the HIV latent reservoir, with a focus on CAR T cell therapies. We highlight the current challenges and advances in this field, while discussing the importance of novel CAR designs in the efforts to find a HIV cure.
Collapse
Affiliation(s)
- Jarrod York
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia
- Centre for Cancer Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia
| | - Kavitha Gowrishankar
- Centre for Cancer Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia
- Children’s Cancer Research Unit, Kids Research, The Children’s Hospital at Westmead, Sydney Children’s Hospitals Network, Westmead, NSW, Australia
- Faculty of Medicine and Health, Sydney Institute for Infectious Diseases, School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
| | - Kenneth Micklethwaite
- Centre for Cancer Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia
- Faculty of Medicine and Health, Sydney Institute for Infectious Diseases, School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
- Blood Transplant and Cell Therapies Program, Department of Haematology, Westmead Hospital, Sydney, NSW, Australia
- NSW Health Pathology Blood Transplant and Cell Therapies Laboratory – Institute of Clinical Pathology and Medical Research (ICPMR) Westmead, Sydney, NSW, Australia
| | - Sarah Palmer
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia
- Faculty of Medicine and Health, Sydney Institute for Infectious Diseases, School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
| | - Anthony L. Cunningham
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia
- Faculty of Medicine and Health, Sydney Institute for Infectious Diseases, School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
| | - Najla Nasr
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, NSW, Australia
- Faculty of Medicine and Health, Sydney Institute for Infectious Diseases, School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
33
|
Constant O, Maarifi G, Blanchet FP, Van de Perre P, Simonin Y, Salinas S. Role of Dendritic Cells in Viral Brain Infections. Front Immunol 2022; 13:862053. [PMID: 35529884 PMCID: PMC9072653 DOI: 10.3389/fimmu.2022.862053] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/22/2022] [Indexed: 11/13/2022] Open
Abstract
To gain access to the brain, a so-called immune-privileged organ due to its physical separation from the blood stream, pathogens and particularly viruses have been selected throughout evolution for their use of specific mechanisms. They can enter the central nervous system through direct infection of nerves or cerebral barriers or through cell-mediated transport. Indeed, peripheral lymphoid and myeloid immune cells can interact with the blood-brain and the blood-cerebrospinal fluid barriers and allow viral brain access using the "Trojan horse" mechanism. Among immune cells, at the frontier between innate and adaptive immune responses, dendritic cells (DCs) can be pathogen carriers, regulate or exacerbate antiviral responses and neuroinflammation, and therefore be involved in viral transmission and spread. In this review, we highlight an important contribution of DCs in the development and the consequences of viral brain infections.
Collapse
Affiliation(s)
- Orianne Constant
- Pathogenesis and Control of Chronic and Emerging Infections, Institut national de la santé et de la recherche médicale (INSERM), University of Montpellier, Etablissement Français du Sang, Montpellier, France
| | - Ghizlane Maarifi
- Institut de Recherche en Infectiologie de Montpellier, Centre national de la recherche scientifique (CNRS), Université de Montpellier, Montpellier, France
| | - Fabien P. Blanchet
- Institut de Recherche en Infectiologie de Montpellier, Centre national de la recherche scientifique (CNRS), Université de Montpellier, Montpellier, France
| | - Philippe Van de Perre
- Pathogenesis and Control of Chronic and Emerging Infections, Institut national de la santé et de la recherche médicale (INSERM), University of Montpellier, Etablissement Français du Sang, Montpellier, France
| | - Yannick Simonin
- Pathogenesis and Control of Chronic and Emerging Infections, Institut national de la santé et de la recherche médicale (INSERM), University of Montpellier, Etablissement Français du Sang, Montpellier, France
| | - Sara Salinas
- Pathogenesis and Control of Chronic and Emerging Infections, Institut national de la santé et de la recherche médicale (INSERM), University of Montpellier, Etablissement Français du Sang, Montpellier, France
| |
Collapse
|
34
|
CGRP inhibits human Langerhans cells infection with HSV by differentially modulating specific HSV-1 and HSV-2 entry mechanisms. Mucosal Immunol 2022; 15:762-771. [PMID: 35562558 DOI: 10.1038/s41385-022-00521-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 04/05/2022] [Accepted: 04/25/2022] [Indexed: 02/04/2023]
Abstract
Herpes simplex virus (HSV) is widespread globally, with both HSV-1 and HSV-2 responsible for genital herpes. During sexual transmission, HSV targets epithelial cells, sensory peripheral pain neurons secreting the mucosal neuropeptide calcitonin gene-related peptide (CGRP), and mucosal immune cells including Langerhans cells (LCs). We previously described a neuro-immune crosstalk, whereby CGRP inhibits LCs-mediated human immunodeficiency virus type 1 (HIV-1) transmission. Herein, to further explore CGRP-mediated anti-viral function, we investigated whether CGRP affects LCs infection with HSV. We found that both HSV-1 and HSV-2 primary isolates productively infect monocyte-derived LCs (MDLCs) and inner foreskin LCs. Moreover, CGRP significantly inhibits infection with both HSV subtypes of MDLCs and langerinhigh, but not langerinlow, inner foreskin LCs. For HSV-1, infection is mediated via the HSV-1-specific entry receptor 3-O sulfated heparan sulfate (3-OS HS) in a pH-depended manner, and CGRP down-regulates 3-OS HS surface expression, as well as abrogates pH dependency. For HSV-2, infection involves langerin-mediated endocytosis in a pH-independent manner, and CGRP up-regulates surface expression of atypical langerin double-trimer oligomers. Our results show that CGRP inhibits mucosal HSV infection by differentially modulating subtype-specific entry receptors and mechanisms in human LCs. CGRP could turn out useful for prevention of LCs-mediated HSV infection and HSV/HIV-1 co-infection.
Collapse
|
35
|
Chinn AM, Salmerón C, Lee J, Sriram K, Raz E, Insel PA. PDE4B Is a Homeostatic Regulator of Cyclic AMP in Dendritic Cells. Front Pharmacol 2022; 13:833832. [PMID: 35387344 PMCID: PMC8977838 DOI: 10.3389/fphar.2022.833832] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 02/01/2022] [Indexed: 11/13/2022] Open
Abstract
Chronic decreases in the second messenger cyclic AMP (cAMP) occur in numerous settings, but how cells compensate for such decreases is unknown. We have used a unique system-murine dendritic cells (DCs) with a DC-selective depletion of the heterotrimeric GTP binding protein Gαs-to address this issue. These mice spontaneously develop Th2-allergic asthma and their DCs have persistently lower cAMP levels. We found that phosphodiesterase 4B (PDE4B) is the primary phosphodiesterase expressed in DCs and that its expression is preferentially decreased in Gαs-depleted DCs. PDE4B expression is dynamic, falling and rising in a protein kinase A-dependent manner with decreased and increased cAMP concentrations, respectively. Treatment of DCs that drive enhanced Th2 immunity with a PDE4B inhibitor ameliorated DC-induced helper T cell response. We conclude that PDE4B is a homeostatic regulator of cellular cAMP concentrations in DCs and may be a target for treating Th2-allergic asthma and other settings with low cellular cAMP concentrations.
Collapse
Affiliation(s)
- Amy M. Chinn
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, United States
| | - Cristina Salmerón
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, United States
| | - Jihyung Lee
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Krishna Sriram
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, United States
| | - Eyal Raz
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Paul A. Insel
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, United States
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
36
|
Bertacchi G, Posch W, Wilflingseder D. HIV-1 Trans Infection via TNTs Is Impeded by Targeting C5aR. Biomolecules 2022; 12:biom12020313. [PMID: 35204813 PMCID: PMC8868603 DOI: 10.3390/biom12020313] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/03/2022] [Accepted: 02/10/2022] [Indexed: 02/04/2023] Open
Abstract
Nonadjacent immune cells communicate through a complex network of tunneling nanotubes (TNTs). TNTs can be hijacked by HIV-1, allowing it to spread between connected cells. Dendritic cells (DCs) are among the first cells to encounter HIV-1 at mucosal sites, but they are usually efficiently infected only at low levels. However, HIV-1 was demonstrated to productively infect DCs when the virus was complement-opsonized (HIV-C). Such HIV-C-exposed DCs mediated an improved antiviral and T-cell stimulatory capacity. The role of TNTs in combination with complement in enhancing DC infection with HIV-C remains to be addressed. To this aim, we evaluated TNT formation on the surface of DCs or DC/CD4+ T-cell co-cultures incubated with non- or complement-opsonized HIV-1 (HIV, HIV-C) and the role of TNTs or locally produced complement in the infection process using either two different TNT or anaphylatoxin receptor antagonists. We found that HIV-C significantly increased the formation of TNTs between DCs or DC/CD4+ T-cell co-cultures compared to HIV-exposed DCs or co-cultures. While augmented TNT formation in DCs promoted productive infection, as was previously observed, a significant reduction in productive infection was observed in DC/CD4+ T-cell co-cultures, indicating antiviral activity in this setting. As expected, TNT inhibitors significantly decreased infection of HIV-C-loaded-DCs as well as HIV- and HIV-C-infected-DC/CD4+ T-cell co-cultures. Moreover, antagonizing C5aR significantly inhibited TNT formation in DCs as well as DC/CD4+ T-cell co-cultures and lowered the already decreased productive infection in co-cultures. Thus, local complement mobilization via DC stimulation of complement receptors plays a pivotal role in TNT formation, and our findings herein might offer an exciting opportunity for novel therapeutic approaches to inhibit trans infection via C5aR targeting.
Collapse
|
37
|
Pampusch MS, Abdelaal HM, Cartwright EK, Molden JS, Davey BC, Sauve JD, Sevcik EN, Rendahl AK, Rakasz EG, Connick E, Berger EA, Skinner PJ. CAR/CXCR5-T cell immunotherapy is safe and potentially efficacious in promoting sustained remission of SIV infection. PLoS Pathog 2022; 18:e1009831. [PMID: 35130312 PMCID: PMC8853520 DOI: 10.1371/journal.ppat.1009831] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 02/17/2022] [Accepted: 01/18/2022] [Indexed: 02/01/2023] Open
Abstract
During chronic human immunodeficiency virus (HIV) or simian immunodeficiency virus (SIV) infection prior to AIDS progression, the vast majority of viral replication is concentrated within B cell follicles of secondary lymphoid tissues. We investigated whether infusion of T cells expressing an SIV-specific chimeric antigen receptor (CAR) and the follicular homing receptor, CXCR5, could successfully kill viral-RNA+ cells in targeted lymphoid follicles in SIV-infected rhesus macaques. In this study, CD4 and CD8 T cells from rhesus macaques were genetically modified to express antiviral CAR and CXCR5 moieties (generating CAR/CXCR5-T cells) and autologously infused into a chronically infected animal. At 2 days post-treatment, the CAR/CXCR5-T cells were located primarily in spleen and lymph nodes both inside and outside of lymphoid follicles. Few CAR/CXCR5-T cells were detected in the ileum, rectum, and lung, and no cells were detected in the bone marrow, liver, or brain. Within follicles, CAR/CXCR5-T cells were found in direct contact with SIV-viral RNA+ cells. We next infused CAR/CXCR5-T cells into ART-suppressed SIV-infected rhesus macaques, in which the animals were released from ART at the time of infusion. These CAR/CXCR5-T cells replicated in vivo within both the extrafollicular and follicular regions of lymph nodes and accumulated within lymphoid follicles. CAR/CXR5-T cell concentrations in follicles peaked during the first week post-infusion but declined to undetectable levels after 2 to 4 weeks. Overall, CAR/CXCR5-T cell-treated animals maintained lower viral loads and follicular viral RNA levels than untreated control animals, and no outstanding adverse reactions were noted. These findings indicate that CAR/CXCR5-T cell treatment is safe and holds promise as a future treatment for the durable remission of HIV.
Collapse
Affiliation(s)
- Mary S. Pampusch
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Hadia M. Abdelaal
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Emily K. Cartwright
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Jhomary S. Molden
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Brianna C. Davey
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Jordan D. Sauve
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Emily N. Sevcik
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Aaron K. Rendahl
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Eva G. Rakasz
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Elizabeth Connick
- Division of Infectious Diseases, University of Arizona, Tucson, Arizona, United States of America
| | - Edward A. Berger
- Laboratory of Viral Diseases, NIAID, NIH, Bethesda, Maryland, United States of America
| | - Pamela J. Skinner
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| |
Collapse
|
38
|
Bourque J, Hawiger D. Applications of Antibody-Based Antigen Delivery Targeted to Dendritic Cells In Vivo. Antibodies (Basel) 2022; 11:antib11010008. [PMID: 35225867 PMCID: PMC8884005 DOI: 10.3390/antib11010008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/11/2022] [Accepted: 01/20/2022] [Indexed: 02/06/2023] Open
Abstract
Recombinant immunoglobulins, derived from monoclonal antibodies recognizing the defined surface epitopes expressed on dendritic cells, have been employed for the past two decades to deliver antigens to dendritic cells in vivo, serving as critical tools for the investigation of the corresponding T cell responses. These approaches originated with the development of the recombinant chimeric antibody against a multilectin receptor, DEC-205, which is present on subsets of murine and human conventional dendritic cells. Following the widespread application of antigen targeting through DEC-205, similar approaches then utilized other epitopes as entry points for antigens delivered by specific antibodies to multiple types of dendritic cells. Overall, these antigen-delivery methodologies helped to reveal the mechanisms underlying tolerogenic and immunogenic T cell responses orchestrated by dendritic cells. Here, we discuss the relevant experimental strategies as well as their future perspectives, including their translational relevance.
Collapse
Affiliation(s)
| | - Daniel Hawiger
- Correspondence: ; Tel.: +1-314-977-8875; Fax: +1-314-977-8717
| |
Collapse
|
39
|
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
|
40
|
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
|
41
|
Mariotton J, Sams A, Cohen E, Sennepin A, Siracusano G, Sanvito F, Edvinsson L, Delongchamps NB, Zerbib M, Lopalco L, Bomsel M, Ganor Y. Native CGRP Neuropeptide and Its Stable Analogue SAX, But Not CGRP Peptide Fragments, Inhibit Mucosal HIV-1 Transmission. Front Immunol 2021; 12:785072. [PMID: 34956215 PMCID: PMC8692891 DOI: 10.3389/fimmu.2021.785072] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 11/16/2021] [Indexed: 11/16/2022] Open
Abstract
Background The vasodilator neuropeptide calcitonin gene-related peptide (CGRP) plays both detrimental and protective roles in different pathologies. CGRP is also an essential component of the neuro-immune dialogue between nociceptors and mucosal immune cells. We previously discovered that CGRP is endowed with anti-viral activity and strongly inhibits human immunodeficiency virus type 1 (HIV-1) infection, by suppressing Langerhans cells (LCs)-mediated HIV-1 trans-infection in-vitro and mucosal HIV-1 transmission ex-vivo. This inhibition is mediated via activation of the CGRP receptor non-canonical NFκB/STAT4 signaling pathway that induces a variety of cooperative mechanisms. These include CGRP-mediated increase in the expression of the LC-specific pathogen recognition C-type lectin langerin and decrease in LC-T-cell conjugates formation. The clinical utility of CGRP and modalities of CGRP receptor activation, for inhibition of mucosal HIV-1 transmission, remain elusive. Methods We tested the capacity of CGRP to inhibit HIV-1 infection in-vivo in humanized mice. We further compared the anti-HIV-1 activities of full-length native CGRP, its metabolically stable analogue SAX, and several CGRP peptide fragments containing its binding C-terminal and activating N-terminal regions. These agonists were evaluated for their capacity to inhibit LCs-mediated HIV-1 trans-infection in-vitro and mucosal HIV-1 transmission in human mucosal tissues ex-vivo. Results A single CGRP intravaginal topical treatment of humanized mice, followed by HIV-1 vaginal challenge, transiently restricts the increase in HIV-1 plasma viral loads but maintains long-lasting higher CD4+ T-cell counts. Similarly to CGRP, SAX inhibits LCs-mediated HIV-1 trans-infection in-vitro, but with lower potency. This inhibition is mediated via CGRP receptor activation, leading to increased expression of both langerin and STAT4 in LCs. In contrast, several N-terminal and N+C-terminal bivalent CGRP peptide fragments fail to increase langerin and STAT4, and accordingly lack anti-HIV-1 activities. Finally, like CGRP, treatment of human inner foreskin tissue explants with SAX, followed by polarized inoculation with cell-associated HIV-1, completely blocks formation of LC-T-cell conjugates and HIV-1 infection of T-cells. Conclusion Our results show that CGRP receptor activation by full-length CGRP or SAX is required for efficient inhibition of LCs-mediated mucosal HIV-1 transmission. These findings suggest that formulations containing CGRP, SAX and/or their optimized agonists/analogues could be harnessed for HIV-1 prevention.
Collapse
Affiliation(s)
- Jammy Mariotton
- Laboratory of Mucosal Entry of HIV-1 and Mucosal Immunity, Department of Infection, Immunity and Inflammation, Institut Cochin, Université de Paris, INSERM U1016, CNRS UMR8104, Paris, France
| | - Anette Sams
- Department of Clinical Experimental Research, Glostrup Research Institute, Copenhagen University Hospital, Copenhagen, Denmark
| | - Emmanuel Cohen
- Laboratory of Mucosal Entry of HIV-1 and Mucosal Immunity, Department of Infection, Immunity and Inflammation, Institut Cochin, Université de Paris, INSERM U1016, CNRS UMR8104, Paris, France
| | - Alexis Sennepin
- Laboratory of Mucosal Entry of HIV-1 and Mucosal Immunity, Department of Infection, Immunity and Inflammation, Institut Cochin, Université de Paris, INSERM U1016, CNRS UMR8104, Paris, France
| | - Gabriel Siracusano
- Emerging Bacterial Pathogens Unit, IRCCS San Raffaele Hospital, Milan, Italy
| | - Francesca Sanvito
- Pathology Unit, Division of Experimental Oncology, IRCCS San Raffaele Hospital, Milan, Italy
| | - Lars Edvinsson
- Department of Clinical Experimental Research, Glostrup Research Institute, Copenhagen University Hospital, Copenhagen, Denmark
| | | | - Marc Zerbib
- Urology Service, GH Cochin-St Vincent de Paul, Paris, France
| | - Lucia Lopalco
- Immunobiology of HIV, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Morgane Bomsel
- Laboratory of Mucosal Entry of HIV-1 and Mucosal Immunity, Department of Infection, Immunity and Inflammation, Institut Cochin, Université de Paris, INSERM U1016, CNRS UMR8104, Paris, France
| | - Yonatan Ganor
- Laboratory of Mucosal Entry of HIV-1 and Mucosal Immunity, Department of Infection, Immunity and Inflammation, Institut Cochin, Université de Paris, INSERM U1016, CNRS UMR8104, Paris, France
| |
Collapse
|
42
|
Tuong ZK, Lukowski SW, Nguyen QH, Chandra J, Zhou C, Gillinder K, Bashaw AA, Ferdinand JR, Stewart BJ, Teoh SM, Hanson SJ, Devitt K, Clatworthy MR, Powell JE, Frazer IH. A model of impaired Langerhans cell maturation associated with HPV induced epithelial hyperplasia. iScience 2021; 24:103326. [PMID: 34805788 PMCID: PMC8586807 DOI: 10.1016/j.isci.2021.103326] [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: 07/09/2021] [Revised: 09/29/2021] [Accepted: 10/19/2021] [Indexed: 12/24/2022] Open
Abstract
Langerhans cells (LC) are skin-resident antigen-presenting cells that regulate immune responses to epithelial microorganisms. Human papillomavirus (HPV) infection can promote malignant epithelial transformation. As LCs are considered important for controlling HPV infection, we compared the transcriptome of murine LCs from skin transformed by K14E7 oncoprotein and from healthy skin. We identified transcriptome heterogeneity at the single cell level amongst LCs in normal skin, associated with ontogeny, cell cycle, and maturation. We identified a balanced co-existence of immune-stimulatory and immune-inhibitory LC cell states in normal skin that was significantly disturbed in HPV16 E7-transformed skin. Hyperplastic skin was depleted of immune-stimulatory LCs and enriched for LCs with an immune-inhibitory gene signature, and LC-keratinocyte crosstalk was dysregulated. We identified reduced expression of interleukin (IL)-34, a critical molecule for LC homeostasis. Enrichment of an immune-inhibitory LC gene signature and reduced levels of epithelial IL-34 were also found in human HPV-associated cervical epithelial cancers. Single cell atlas of Langerhans cells in cutaneous skin Stimulatory and inhibitory Langerhans cell states are in balance Inhibitory Langerhans cell states dominate HPV-transformed hyperplastic skin Langerhans cell imbalance is associated with disrupted IL-34 signaling
Collapse
Affiliation(s)
- Zewen K Tuong
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia.,Molecular Immunity Unit, University of Cambridge Department of Medicine, MRC-Laboratory of Molecular Biology, Cambridge, UK
| | - Samuel W Lukowski
- Australia Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Quan H Nguyen
- Australia Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Janin Chandra
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Chenhao Zhou
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Kevin Gillinder
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Abate A Bashaw
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - John R Ferdinand
- Molecular Immunity Unit, University of Cambridge Department of Medicine, MRC-Laboratory of Molecular Biology, Cambridge, UK
| | - Benjamin J Stewart
- Molecular Immunity Unit, University of Cambridge Department of Medicine, MRC-Laboratory of Molecular Biology, Cambridge, UK
| | - Siok Min Teoh
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Sarah J Hanson
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Katharina Devitt
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Menna R Clatworthy
- Molecular Immunity Unit, University of Cambridge Department of Medicine, MRC-Laboratory of Molecular Biology, Cambridge, UK.,Wellcome Trust Sanger Institute, Hinxton, UK
| | - Joseph E Powell
- Garvan-Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - Ian H Frazer
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| |
Collapse
|
43
|
Giza HM, Bozzacco L. Unboxing dendritic cells: Tales of multi-faceted biology and function. Immunology 2021; 164:433-449. [PMID: 34309853 PMCID: PMC8517577 DOI: 10.1111/imm.13394] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/13/2021] [Accepted: 07/14/2021] [Indexed: 12/14/2022] Open
Abstract
Often referred to as the bridge between innate and adaptive immunity, dendritic cells (DCs) are professional antigen-presenting cells (APCs) that constitute a unique, yet complex cell system. Among other APCs, DCs display the unique property of inducing protective immune responses against invading microbes, or cancer cells, while safeguarding the proper homeostatic equilibrium of the immune system and maintaining self-tolerance. Unsurprisingly, DCs play a role in many diseases such as autoimmunity, allergy, infectious disease and cancer. This makes them attractive but challenging targets for therapeutics. Since their initial discovery, research and understanding of DC biology have flourished. We now recognize the presence of multiple subsets of DCs distributed across tissues. Recent studies of phenotype and gene expression at the single cell level have identified heterogeneity even within the same DC type, supporting the idea that DCs have evolved to greatly expand the flexibility of the immune system to react appropriately to a wide range of threats. This review is meant to serve as a quick and robust guide to understand the basic divisions of DC subsets and their role in the immune system. Between mice and humans, there are some differences in how these subsets are identified and function, and we will point out specific distinctions as necessary. Throughout the text, we are using both fundamental and therapeutic lens to describe overlaps and distinctions and what this could mean for future research and therapies.
Collapse
|
44
|
How dendritic cells sense and respond to viral infections. Clin Sci (Lond) 2021; 135:2217-2242. [PMID: 34623425 DOI: 10.1042/cs20210577] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 09/15/2021] [Accepted: 09/23/2021] [Indexed: 12/26/2022]
Abstract
The ability of dendritic cells (DCs) to sense viral pathogens and orchestrate a proper immune response makes them one of the key players in antiviral immunity. Different DC subsets have complementing functions during viral infections, some specialize in antigen presentation and cross-presentation and others in the production of cytokines with antiviral activity, such as type I interferons. In this review, we summarize the latest updates concerning the role of DCs in viral infections, with particular focus on the complex interplay between DC subsets and severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). Despite being initiated by a vast array of immune receptors, DC-mediated antiviral responses often converge towards the same endpoint, that is the production of proinflammatory cytokines and the activation of an adaptive immune response. Nonetheless, the inherent migratory properties of DCs make them a double-edged sword and often viral recognition by DCs results in further viral dissemination. Here we illustrate these various aspects of the antiviral functions of DCs and also provide a brief overview of novel antiviral vaccination strategies based on DCs targeting.
Collapse
|
45
|
Abstract
The concept of functional specialization is fundamental to the immune system but has not been previously observed in human Langerhans cells. In this issue of Immunity, Liu et al. use single-cell approaches to define two distinct epidermal subsets converging on a common activation and migration pathway.
Collapse
Affiliation(s)
- Matthew Collin
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; Northern Centre for Cancer Care, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE7 7DN, UK
| | - Venetia Bigley
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; Northern Centre for Cancer Care, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE7 7DN, UK.
| |
Collapse
|
46
|
Freen-van Heeren JJ. Flow-FISH as a Tool for Studying Bacteria, Fungi and Viruses. BIOTECH 2021; 10:21. [PMID: 35822795 PMCID: PMC9245478 DOI: 10.3390/biotech10040021] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 10/03/2021] [Accepted: 10/08/2021] [Indexed: 12/20/2022] Open
Abstract
Many techniques are currently in use to study microbes. These can be aimed at detecting, identifying, and characterizing bacterial, fungal, and viral species. One technique that is suitable for high-throughput analysis is flow cytometry-based fluorescence in situ hybridization, or Flow-FISH. This technique employs (fluorescently labeled) probes directed against DNA or (m)RNA, for instance targeting a gene or microorganism of interest and provides information on a single-cell level. Furthermore, by combining Flow-FISH with antibody-based protein detection, proteins of interest can be measured simultaneously with genetic material. Additionally, depending on the type of Flow-FISH assay, Flow-FISH can also be multiplexed, allowing for the simultaneous measurement of multiple gene targets and/or microorganisms. Together, this allows for, e.g., single-cell gene expression analysis or identification of (sub)strains in mixed cultures. Flow-FISH has been used in mammalian cells but has also been extensively employed to study diverse microbial species. Here, the use of Flow-FISH for studying microorganisms is reviewed. Specifically, the detection of (intracellular) pathogens, studying microorganism biology and disease pathogenesis, and identification of bacterial, fungal, and viral strains in mixed cultures is discussed, with a particular focus on the viruses EBV, HIV-1, and SARS-CoV-2.
Collapse
|
47
|
Wang T, Zhou Z, Luo E, Zhong J, Zhao D, Dong H, Yao B. Comprehensive RNA sequencing in primary murine keratinocytes and fibroblasts identifies novel biomarkers and provides potential therapeutic targets for skin-related diseases. Cell Mol Biol Lett 2021; 26:42. [PMID: 34602061 PMCID: PMC8489068 DOI: 10.1186/s11658-021-00285-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 09/24/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Keratinocytes and fibroblasts represent the major cell types in the epidermis and dermis of the skin and play a significant role in maintenance of skin homeostasis. However, the biological characteristics of keratinocytes and fibroblasts remain to be elucidated. The purpose of this study was to compare the gene expression pattern between keratinocytes and fibroblasts and to explore novel biomarker genes so as to provide potential therapeutic targets for skin-related diseases such as burns, wounds, and aging. METHODS Skin keratinocytes and fibroblasts were isolated from newborn mice. To fully understand the heterogeneity of gene expression between keratinocytes and fibroblasts, differentially expressed genes (DEGs) between the two cell types were detected by RNA-seq technology. Quantitative real-time polymerase chain reaction (qRT-PCR) was used to detect the known genes of keratinocytes and fibroblasts and verify the RNA-seq results. RESULTS Transcriptomic data showed a total of 4309 DEGs (fold-change > 1.5 and q-value < 0.05). Among them, 2197 genes were highly expressed in fibroblasts and included 10 genes encoding collagen, 16 genes encoding transcription factors, and 14 genes encoding growth factors. Simultaneously, 2112 genes were highly expressed in keratinocytes and included 7 genes encoding collagen, 14 genes encoding transcription factors, and 8 genes encoding growth factors. Furthermore, we summarized 279 genes specifically expressed in keratinocytes and 33 genes specifically expressed in fibroblasts, which may represent distinct molecular signatures of each cell type. Additionally, we observed some novel specific biomarkers for fibroblasts such as Plac8 (placenta-specific 8), Agtr2 (angiotensin II receptor, type 2), Serping1 (serpin peptidase inhibitor, clade G, member 1), Ly6c1 (lymphocyte antigen 6 complex, locus C1), Dpt (dermatopontin), and some novel specific biomarkers for keratinocytes such as Ly6a (lymphocyte antigen 6 complex, locus A) and Lce3c (late cornified envelope 3C), Ccer2 (coiled-coil glutamate-rich protein 2), Col18a1 (collagen, type XVIII, alpha 1) and Col17a1 (collagen type XVII, alpha 1). In summary, these data provided novel identifying biomarkers for two cell types, which can provide a resource of DEGs for further investigations.
Collapse
Affiliation(s)
- Tiancheng Wang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Zhenwei Zhou
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Enjing Luo
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Jinghong Zhong
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Daqing Zhao
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Haisi Dong
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117, China.
| | - Baojin Yao
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117, China.
| |
Collapse
|
48
|
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
|
49
|
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
|
50
|
Nijmeijer BM, Langedijk CJM, Geijtenbeek TBH. Mucosal Dendritic Cell Subsets Control HIV-1's Viral Fitness. Annu Rev Virol 2021; 7:385-402. [PMID: 32991263 DOI: 10.1146/annurev-virology-020520-025625] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Dendritic cell (DC) subsets are abundantly present in genital and intestinal mucosal tissue and are among the first innate immune cells that encounter human immunodeficiency virus type 1 (HIV-1) after sexual contact. Although DCs have specific characteristics that greatly enhance HIV-1 transmission, it is becoming evident that most DC subsets also have virus restriction mechanisms that exert selective pressure on the viruses during sexual transmission. In this review we discuss the current concepts of the immediate events following viral exposure at genital mucosal sites that lead to selection of specific HIV-1 variants called transmitted founder (TF) viruses. We highlight the importance of the TF HIV-1 phenotype and the role of different DC subsets in establishing infection. Understanding the biology of HIV-1 transmission will contribute to the design of novel treatment strategies preventing HIV-1 dissemination.
Collapse
Affiliation(s)
- Bernadien M Nijmeijer
- Department of Experimental Immunology, Amsterdam Institute of Infection and Immunity, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
| | - Catharina J M Langedijk
- Department of Experimental Immunology, Amsterdam Institute of Infection and Immunity, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
| | - Teunis B H Geijtenbeek
- Department of Experimental Immunology, Amsterdam Institute of Infection and Immunity, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
| |
Collapse
|