1
|
Kotlyarov S, Oskin D. The Role of Inflammation in the Pathogenesis of Comorbidity of Chronic Obstructive Pulmonary Disease and Pulmonary Tuberculosis. Int J Mol Sci 2025; 26:2378. [PMID: 40141021 PMCID: PMC11942565 DOI: 10.3390/ijms26062378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 02/23/2025] [Accepted: 03/05/2025] [Indexed: 03/28/2025] Open
Abstract
The comorbid course of chronic obstructive pulmonary disease (COPD) and pulmonary tuberculosis is an important medical and social problem. Both diseases, although having different etiologies, have many overlapping relationships that mutually influence their course and prognosis. The aim of the current review is to discuss the role of different immune mechanisms underlying inflammation in COPD and pulmonary tuberculosis. These mechanisms are known to involve both the innate and adaptive immune system, including various cellular and intercellular interactions. There is growing evidence that immune mechanisms involved in the pathogenesis of both COPD and tuberculosis may jointly contribute to the tuberculosis-associated obstructive pulmonary disease (TOPD) phenotype. Several studies have reported prior tuberculosis as a risk factor for COPD. Therefore, the study of the mechanisms that link COPD and tuberculosis is of considerable clinical interest.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| | - Dmitry Oskin
- Department of Infectious Diseases and Phthisiology, Ryazan State Medical University, 390026 Ryazan, Russia
| |
Collapse
|
2
|
Lukeman H, Al-Wassiti H, Fabb SA, Lim L, Wang T, Britton WJ, Steain M, Pouton CW, Triccas JA, Counoupas C. An LNP-mRNA vaccine modulates innate cell trafficking and promotes polyfunctional Th1 CD4 + T cell responses to enhance BCG-induced protective immunity against Mycobacterium tuberculosis. EBioMedicine 2025; 113:105599. [PMID: 39955975 PMCID: PMC11871481 DOI: 10.1016/j.ebiom.2025.105599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 01/28/2025] [Accepted: 01/31/2025] [Indexed: 02/18/2025] Open
Abstract
BACKGROUND Mycobacterium tuberculosis remains the largest infectious cause of mortality worldwide, even with over a century of widespread administration of the only licenced tuberculosis (TB) vaccine, Bacillus Calmette-Guérin (BCG). mRNA technology remains an underexplored approach for combating chronic bacterial infections such as TB. METHODS We have developed a lipid nanoparticle (LNP)-mRNA vaccine, termed mRNACV2, encoding for the M. tuberculosis CysVac2 fusion protein, which we have previously formulated as an adjuvanted subunit vaccine. This LNP-mRNA vaccine was administered intramuscularly to female C57BL/6 mice as a standalone vaccine or as booster to BCG to assess immunogenicity and efficacy of the construct. FINDINGS Vaccination with mRNACV2 induced high frequencies of polyfunctional, antigen-specific Th1 CD4+ T cells in the blood and lungs, which was associated with the rapid recruitment of both innate and adaptive immune cells to lymph nodes draining the site of immunisation. mRNACV2 vaccination also provided significant pulmonary protection in M. tuberculosis-infected mice, reducing bacterial load and inflammatory infiltration in the lungs. Importantly, mRNACV2 enhanced immune responses and long-term protection when used to boost BCG-primed mice. INTERPRETATION These findings of a protective LNP-mRNA vaccine for TB highlight the potential of the LNP-mRNA platform for TB control and support further research to facilitate translation to humans. FUNDING This work was supported by the NHMRC Centre of Research Excellence in Tuberculosis Control to JAT and WJB (APP1153493), and MRFF mRNA Clinical Trial Enabling Infrastructure grant to CWP and HAW (MRFCTI000006).
Collapse
Affiliation(s)
- Hannah Lukeman
- Sydney Infectious Diseases Institute (Sydney ID) and School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, 2006, Australia; Centre for Infection and Immunity, Centenary Institute, The University of Sydney, Camperdown, NSW, 2006, Australia; Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Hareth Al-Wassiti
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, 3052, Australia
| | - Stewart A Fabb
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, 3052, Australia
| | - Leonard Lim
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, 3052, Australia
| | - Trixie Wang
- Centre for Infection and Immunity, Centenary Institute, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Warwick J Britton
- Centre for Infection and Immunity, Centenary Institute, The University of Sydney, Camperdown, NSW, 2006, Australia; Department of Clinical Immunology, Royal Prince Alfred Hospital, Camperdown, NSW, 2050, Australia
| | - Megan Steain
- Sydney Infectious Diseases Institute (Sydney ID) and School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, 2006, Australia; Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Colin W Pouton
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, 3052, Australia
| | - James A Triccas
- Sydney Infectious Diseases Institute (Sydney ID) and School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, 2006, Australia; Centre for Infection and Immunity, Centenary Institute, The University of Sydney, Camperdown, NSW, 2006, Australia; Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia.
| | - Claudio Counoupas
- Sydney Infectious Diseases Institute (Sydney ID) and School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, 2006, Australia; Centre for Infection and Immunity, Centenary Institute, The University of Sydney, Camperdown, NSW, 2006, Australia; Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia.
| |
Collapse
|
3
|
Dudziak D, Heger L, Agace WW, Bakker J, de Gruijl TD, Dress RJ, Dutertre C, Fenton TM, Fransen MF, Ginhoux F, Heyman O, Horev Y, Hornsteiner F, Kandiah V, Kles P, Lubin R, Mizraji G, Prokopi A, Saar O, Sopper S, Stoitzner P, Strandt H, Sykora MM, Toffoli EC, Tripp CH, van Pul K, van de Ven R, Wilensky A, Yona S, Zelle‐Rieser C. Guidelines for preparation and flow cytometry analysis of human nonlymphoid tissue DC. Eur J Immunol 2025; 55:e2250325. [PMID: 39668411 PMCID: PMC11739683 DOI: 10.1002/eji.202250325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 09/19/2024] [Accepted: 09/25/2024] [Indexed: 12/14/2024]
Abstract
This article is part of the Dendritic Cell Guidelines article series, which provides a collection of state-of-the-art protocols for the preparation, phenotype analysis by flow cytometry, generation, fluorescence microscopy, and functional characterization of mouse and human dendritic cells (DC) from lymphoid organs, and various nonlymphoid tissues. Within this article, detailed protocols are presented that allow for the generation of single-cell suspensions from human nonlymphoid tissues including lung, skin, gingiva, intestine as well as from tumors and tumor-draining lymph nodes with a subsequent analysis of dendritic cells by flow cytometry. Further, prepared single-cell suspensions can be subjected to other applications including cellular enrichment procedures, RNA sequencing, functional assays, etc. While all protocols were written by experienced scientists who routinely use them in their work, this article was also peer-reviewed by leading experts and approved by all co-authors, making it an essential resource for basic and clinical DC immunologists.
Collapse
Affiliation(s)
- Diana Dudziak
- Institute of ImmunologyJena University HospitalFriedrich‐Schiller‐UniversityJenaGermany
- Laboratory of Dendritic Cell BiologyDepartment of DermatologyUniversity Hospital ErlangenErlangenGermany
| | - Lukas Heger
- Laboratory of Dendritic Cell BiologyDepartment of DermatologyUniversity Hospital ErlangenErlangenGermany
- Department of Transfusion Medicine and HemostaseologyUniversity Hospital ErlangenErlangenGermany
| | - William W Agace
- LEO Foundation Skin Immunology Research CenterDepartment of Immunology and MicrobiologyUniversity of CopenhagenCopenhagenDenmark
- Immunology SectionLund UniversityLundSweden
| | - Joyce Bakker
- Institute for Infection and ImmunologyCancer ImmunologyAmsterdamThe Netherlands
- Cancer Center AmsterdamCancer ImmunologyAmsterdamThe Netherlands
- Amsterdam UMC location Vrije UniversiteitMedical OncologyAmsterdamThe Netherlands
| | - Tanja D. de Gruijl
- Institute for Infection and ImmunologyCancer ImmunologyAmsterdamThe Netherlands
- Cancer Center AmsterdamCancer ImmunologyAmsterdamThe Netherlands
- Amsterdam UMC location Vrije UniversiteitMedical OncologyAmsterdamThe Netherlands
| | - Regine J. Dress
- Institute of Systems ImmunologyHamburg Center for Translational Immunology (HCTI)University Medical Center Hamburg‐EppendorfHamburgGermany
| | | | | | - Marieke F. Fransen
- Institute for Infection and ImmunologyCancer ImmunologyAmsterdamThe Netherlands
- Cancer Center AmsterdamCancer ImmunologyAmsterdamThe Netherlands
- Department of Pulmonary DiseasesAmsterdam UMC location Vrije UniversiteitAmsterdamThe Netherlands
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and ResearchSingaporeSingapore
- Department of Immunology and MicrobiologyShanghai Institute of ImmunologyShanghai Jiao Tong University School of MedicineShanghaiChina
- SingHealth Duke‐NUS Academic Medical CentreTranslational Immunology InstituteSingaporeSingapore
- INSERM U1015, Gustave Roussy Cancer CampusVillejuifFrance
| | - Oded Heyman
- Department of PeriodontologyHadassah Medical CenterFaculty of Dental MedicineHebrew University of JerusalemIsrael
| | - Yael Horev
- Department of PeriodontologyHadassah Medical CenterFaculty of Dental MedicineHebrew University of JerusalemIsrael
| | - Florian Hornsteiner
- Department of Dermatology, Venereology & AllergologyMedical University of InnsbruckInnsbruckAustria
| | - Vinitha Kandiah
- Institute for Infection and ImmunologyCancer ImmunologyAmsterdamThe Netherlands
- Cancer Center AmsterdamCancer ImmunologyAmsterdamThe Netherlands
- Amsterdam UMC location Vrije UniversiteitMedical OncologyAmsterdamThe Netherlands
| | - Paz Kles
- Department of PeriodontologyHadassah Medical CenterFaculty of Dental MedicineHebrew University of JerusalemIsrael
| | - Ruth Lubin
- Faculty of Dental MedicineThe Institute of Biomedical and Oral ResearchHebrew University of JerusalemIsrael
| | - Gabriel Mizraji
- Department of PeriodontologyHadassah Medical CenterFaculty of Dental MedicineHebrew University of JerusalemIsrael
| | - Anastasia Prokopi
- Institute for Infection and ImmunologyCancer ImmunologyAmsterdamThe Netherlands
- Cancer Center AmsterdamCancer ImmunologyAmsterdamThe Netherlands
- Amsterdam UMC location Vrije UniversiteitMedical OncologyAmsterdamThe Netherlands
| | - Or Saar
- Department of PeriodontologyHadassah Medical CenterFaculty of Dental MedicineHebrew University of JerusalemIsrael
| | - Sieghart Sopper
- Internal Medicine V, Hematology and OncologyMedical University of InnsbruckInnsbruckAustria
- Tyrolean Cancer Research CenterInnsbruckAustria
| | - Patrizia Stoitzner
- Department of Dermatology, Venereology & AllergologyMedical University of InnsbruckInnsbruckAustria
| | - Helen Strandt
- Department of Dermatology, Venereology & AllergologyMedical University of InnsbruckInnsbruckAustria
| | - Martina M Sykora
- Internal Medicine V, Hematology and OncologyMedical University of InnsbruckInnsbruckAustria
- Tyrolean Cancer Research CenterInnsbruckAustria
| | - Elisa C. Toffoli
- Institute for Infection and ImmunologyCancer ImmunologyAmsterdamThe Netherlands
- Cancer Center AmsterdamCancer ImmunologyAmsterdamThe Netherlands
- Amsterdam UMC location Vrije UniversiteitMedical OncologyAmsterdamThe Netherlands
| | - Christoph H. Tripp
- Department of Dermatology, Venereology & AllergologyMedical University of InnsbruckInnsbruckAustria
| | - Kim van Pul
- Institute for Infection and ImmunologyCancer ImmunologyAmsterdamThe Netherlands
- Cancer Center AmsterdamCancer ImmunologyAmsterdamThe Netherlands
- Amsterdam UMC location Vrije UniversiteitMedical OncologyAmsterdamThe Netherlands
| | - Rieneke van de Ven
- Cancer Center AmsterdamCancer ImmunologyAmsterdamThe Netherlands
- Amsterdam UMC location Vrije UniversiteitMedical OncologyAmsterdamThe Netherlands
- Department of Otolaryngology, Head and Neck SurgeryAmsterdam UMC location Vrije UniversiteitAmsterdamThe Netherlands
| | - Asaf Wilensky
- Department of PeriodontologyHadassah Medical CenterFaculty of Dental MedicineHebrew University of JerusalemIsrael
| | - Simon Yona
- Faculty of Dental MedicineThe Institute of Biomedical and Oral ResearchHebrew University of JerusalemIsrael
| | - Claudia Zelle‐Rieser
- Department of Dermatology, Venereology & AllergologyMedical University of InnsbruckInnsbruckAustria
| |
Collapse
|
4
|
Dinis M, Tran NC. Oral immune system and microbes. MICROBES, MICROBIAL METABOLISM, AND MUCOSAL IMMUNITY 2024:147-228. [DOI: 10.1016/b978-0-323-90144-4.00005-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
5
|
Si Y, Wang Y, Tian Q, Wang Q, Pollard JM, Srivastava PK, Esser-Kahn AP, Collier JH, Sperling AI, Chong AS. Lung cDC1 and cDC2 dendritic cells priming naive CD8 + T cells in situ prior to migration to draining lymph nodes. Cell Rep 2023; 42:113299. [PMID: 37864794 PMCID: PMC10676754 DOI: 10.1016/j.celrep.2023.113299] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 08/21/2023] [Accepted: 10/02/2023] [Indexed: 10/23/2023] Open
Abstract
The current paradigm indicates that naive T cells are primed in secondary lymphoid organs. Here, we present evidence that intranasal administration of peptide antigens appended to nanofibers primes naive CD8+ T cells in the lung independently and prior to priming in the draining mediastinal lymph node (MLN). Notably, comparable accumulation and transcriptomic responses of CD8+ T cells in lung and MLN are observed in both Batf3KO and wild-type (WT) mice, indicating that, while cDC1 dendritic cells (DCs) are the major subset for cross-presentation, cDC2 DCs alone are capable of cross-priming CD8+ T cells both in the lung and draining MLN. Transcription analyses reveal distinct transcriptional responses in lung cDC1 and cDC2 to intranasal nanofiber immunization. However, both DC subsets acquire shared transcriptional responses upon migration into the lymph node, thus uncovering a stepwise activation process of cDC1 and cDC2 toward their ability to cross-prime effector and functional memory CD8+ T cell responses.
Collapse
Affiliation(s)
- Youhui Si
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; Department of Surgery, The University of Chicago, Chicago, IL 60637, USA.
| | - Yihan Wang
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA
| | - Qiaomu Tian
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA
| | - Qiang Wang
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA
| | - Jared M Pollard
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA
| | - Pramod K Srivastava
- Department of Immunology and Carole and Ray Neag Comprehensive Cancer Center, University of Connecticut School of Medicine, Farmington, CT 06032, USA
| | - Aaron P Esser-Kahn
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL 60637, USA
| | - Joel H Collier
- Department of Biomedical Engineering, Duke University, Durham, NC 27710, USA
| | - Anne I Sperling
- Department of Medicine, Pulmonary and Critical Care, University of Virginia, Charlottesville, VA 22908, USA
| | - Anita S Chong
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
6
|
Engel S, Bachem A, Strugnell RA, Strasser A, Herold MJ, Bedoui S. Functional flexibility and plasticity in immune control of systemic Salmonella infection. Curr Opin Immunol 2023; 83:102343. [PMID: 37245415 DOI: 10.1016/j.coi.2023.102343] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/21/2023] [Accepted: 04/30/2023] [Indexed: 05/30/2023]
Abstract
Immunity to systemic Salmonella infection depends on multiple effector mechanisms. Lymphocyte-derived interferon gamma (IFN-γ) enhances cell-intrinsic bactericidal capabilities to antagonize the hijacking of phagocytes as replicative niches for Salmonella. Programmed cell death (PCD) provides another means through which phagocytes fight against intracellular Salmonella. We describe remarkable levels of flexibility with which the host coordinates and adapts these responses. This involves interchangeable cellular sources of IFN-γ regulated by innate and adaptive cues, and the rewiring of PCD pathways in previously unknown ways. We discuss that such plasticity is likely the consequence of host-pathogen coevolution and raise the possibility of further functional overlap between these seemingly distinct processes.
Collapse
Affiliation(s)
- Sven Engel
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, Australia
| | - Annabell Bachem
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, Australia
| | - Richard A Strugnell
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, Australia
| | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia; Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Marco J Herold
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia; Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Sammy Bedoui
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
7
|
Jirmo AC, Grychtol R, Gaedcke S, Liu B, DeStefano S, Happle C, Halle O, Monteiro JT, Habener A, Breiholz OD, DeLuca D, Hansen G. Single cell RNA sequencing reveals distinct clusters of Irf8-expressing pulmonary conventional dendritic cells. Front Immunol 2023; 14:1127485. [PMID: 37251386 PMCID: PMC10213693 DOI: 10.3389/fimmu.2023.1127485] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 04/25/2023] [Indexed: 05/31/2023] Open
Abstract
A single population of interferon-regulatory factor 8 (Irf8)-dependent conventional dendritic cell (cDC type1) is considered to be responsible for both immunogenic and tolerogenic responses depending on the surrounding cytokine milieu. Here, we challenge this concept of an omnipotent single Irf8-dependent cDC1 cluster through analysis of pulmonary cDCs at single cell resolution. We report existence of a pulmonary cDC1 cluster lacking Xcr1 with an immunogenic signature that clearly differs from the Xcr1 positive cDC1 cluster. The Irf8+Batf3+Xcr1- cluster expresses high levels of pro-inflammatory genes associated with antigen presentation, migration and co-stimulation such as Ccr7, Cd74, MHC-II, Ccl5, Il12b and Relb while, the Xcr1+ cDC1 cluster expresses genes corresponding to immune tolerance mechanisms like Clec9a, Pbx1, Cadm1, Btla and Clec12a. In concordance with their pro-inflammatory gene expression profile, the ratio of Xcr1- cDC1s but not Xcr1+cDC1 is increased in the lungs of allergen-treated mice compared to the control group, in which both cDC1 clusters are present in comparable ratios. The existence of two distinct Xcr1+ and Xcr1- cDC1 clusters is furthermore supported by velocity analysis showing markedly different temporal patterns of Xcr1- and Xcr1+cDC1s. In summary, we present evidence for the existence of two different cDC1 clusters with distinct immunogenic profiles in vivo. Our findings have important implications for DC-targeting immunomodulatory therapies.
Collapse
Affiliation(s)
- Adan Chari Jirmo
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Ruth Grychtol
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Svenja Gaedcke
- Biomedical Research in Endstage and Obstructive Lung Disease Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Bin Liu
- Biomedical Research in Endstage and Obstructive Lung Disease Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Stephanie DeStefano
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Christine Happle
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Olga Halle
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Joao T. Monteiro
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
- Excellence Cluster Resolving Infection Susceptibility RESIST (EXC 2155), Deutsche Forschungsgemeinschaft, Hannover Medical School, Hannover, Germany
| | - Anika Habener
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Oliver D. Breiholz
- Research Core Unit Genomics (RCUG), Hannover Medical School, Hannover, Germany
| | - David DeLuca
- Biomedical Research in Endstage and Obstructive Lung Disease Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Gesine Hansen
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
- Excellence Cluster Resolving Infection Susceptibility RESIST (EXC 2155), Deutsche Forschungsgemeinschaft, Hannover Medical School, Hannover, Germany
| |
Collapse
|
8
|
Saraav I, Sibley LD. Dendritic Cells and Cryptosporidium: From Recognition to Restriction. Microorganisms 2023; 11:1056. [PMID: 37110479 PMCID: PMC10144555 DOI: 10.3390/microorganisms11041056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 03/29/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Host immune responses are required for the efficient control of cryptosporidiosis. Immunity against Cryptosporidium infection has been best studied in mice, where it is mediated by both innate and adaptive immune responses. Dendritic cells are the key link between innate and adaptive immunity and participate in the defense against Cryptosporidium infection. While the effector mechanism varies, both humans and mice rely on dendritic cells for sensing parasites and restricting infection. Recently, the use of mouse-adapted strains C. parvum and mouse-specific strain C. tyzzeri have provided tractable systems to study the role of dendritic cells in mice against this parasite. In this review, we provide an overview of recent advances in innate immunity acting during infection with Cryptosporidium with a major focus on the role of dendritic cells in the intestinal mucosa. Further work is required to understand the role of dendritic cells in the activation of T cells and to explore associated molecular mechanisms. The identification of Cryptosporidium antigen involved in the activation of Toll-like receptor signaling in dendritic cells during infection is also a matter of future study. The in-depth knowledge of immune responses in cryptosporidiosis will help develop targeted prophylactic and therapeutic interventions.
Collapse
Affiliation(s)
| | - L. David Sibley
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
9
|
Filardy AA, Ferreira JRM, Rezende RM, Kelsall BL, Oliveira RP. The intestinal microenvironment shapes macrophage and dendritic cell identity and function. Immunol Lett 2023; 253:41-53. [PMID: 36623708 PMCID: PMC9907447 DOI: 10.1016/j.imlet.2023.01.003] [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: 04/29/2022] [Revised: 12/12/2022] [Accepted: 01/04/2023] [Indexed: 01/09/2023]
Abstract
The gut comprises the largest body interface with the environment and is continuously exposed to nutrients, food antigens, and commensal microbes, as well as to harmful pathogens. Subsets of both macrophages and dendritic cells (DCs) are present throughout the intestinal tract, where they primarily inhabit the gut-associate lymphoid tissue (GALT), such as Peyer's patches and isolated lymphoid follicles. In addition to their role in taking up and presenting antigens, macrophages and DCs possess extensive functional plasticity and these cells play complementary roles in maintaining immune homeostasis in the gut by preventing aberrant immune responses to harmless antigens and microbes and by promoting host defense against pathogens. The ability of macrophages and DCs to induce either inflammation or tolerance is partially lineage imprinted, but can also be dictated by their activation state, which in turn is determined by their specific microenvironment. These cells express several surface and intracellular receptors that detect danger signals, nutrients, and hormones, which can affect their activation state. DCs and macrophages play a fundamental role in regulating T cells and their effector functions. Thus, modulation of intestinal mucosa immunity by targeting antigen presenting cells can provide a promising approach for controlling pathological inflammation. In this review, we provide an overview on the characteristics, functions, and origins of intestinal macrophages and DCs, highlighting the intestinal microenvironmental factors that influence their functions during homeostasis. Unraveling the mechanisms by which macrophages and DCs regulate intestinal immunity will deepen our understanding on how the immune system integrates endogenous and exogenous signals in order to maintain the host's homeostasis.
Collapse
Affiliation(s)
- Alessandra A Filardy
- Laboratório de Imunologia Celular, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Brazil.
| | - Jesuino R M Ferreira
- Laboratório de Imunologia Celular, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Brazil
| | - Rafael M Rezende
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, USA
| | - Brian L Kelsall
- Laboratory of Molecular Immunology, NIAID, National Institutes of Health, USA
| | | |
Collapse
|
10
|
Zhuang L, Wang Y, Chen Z, Li Z, Wang Z, Jia K, Zhao J, Zhang H, Xie H, Lu L, Chen K, Chen L, Fukuda K, Sano M, Zhang R, Liu J, Yan X. Global Characteristics and Dynamics of Single Immune Cells After Myocardial Infarction. J Am Heart Assoc 2022; 11:e027228. [PMID: 36515244 PMCID: PMC9798793 DOI: 10.1161/jaha.122.027228] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background Myocardial infarction (MI) is characterized by the emergence of dead or dying cardiomyocytes and excessive immune cell infiltration after coronary vessel occlusion. However, the complex transcriptional profile, pathways, cellular interactome, and transcriptional regulators of immune subpopulations after MI remain elusive. Methods and Results Here, male C57BL/6 mice were subjected to MI surgery and monitored for 1 day and 7 days, or sham surgery for 7 days, then cardiac CD45-positive immune cells were collected for single-cell RNA sequencing to determine immune heterogeneity. A total of 30 135 CD45+ immune cells were partitioned into macrophages, monocytes, neutrophils, dendritic cells, and T or B cells for further analysis. We showed that macrophages enriched for Olr1 and differentially expressed Gpnmb represented 2 crucial ischemia-associated macrophages with distinct proinflammatory and prophagocytic capabilities. In contrast to the proinflammatory subset of macrophages enriched for Olr1, Gpnmb-positive macrophages exhibited higher phagocytosis and fatty acid oxidation preference, which could be abolished by etomoxir treatment. In addition to macrophages, MI triggered prompt recruitment of neutrophils into murine hearts, which constituted the sequential cell-fate from naïve S100a4-positive, to activated Sell-high, to aging Icam1-high neutrophils. In silico tools predicted that the excessively expanded neutrophils at 1 day were attributed to chemokine C-C motif ligand/chemokine C-X-C motif ligand pathways, whereas CD80/inducible T-cell costimulator (ICOS) signaling was responsible for the immunosuppressive response at day 7 after MI. Finally, the Fos/AP-1 (activator protein 1) regulon was identified as the critical regulator of proinflammatory responses, which was significantly activated in patients with dilated cardiomyopathy and ischemic cardiomyopathy. We showed the enriched Fos/AP-1 target gene loci in genome-wide association study signals for coronary artery diseases and MI. Targeting Fos/AP-1 with the selective inhibitor T5224 blunted leukocyte infiltration and alleviated cardiac dysfunction in the preclinical murine MI model. Conclusions Taken together, this single-cell RNA sequencing data lay the groundwork for the understanding of immune cell heterogeneity and dynamics in murine ischemic hearts. Moreover, Fos/AP-1 inhibition mitigates inflammatory responses and cardiac dysfunction, which might provide potential therapeutic benefits for heart failure intervention after MI.
Collapse
Affiliation(s)
- Lingfang Zhuang
- Department of Cardiovascular Medicine, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiPR China,Institute of Cardiovascular DiseasesShanghai Jiao Tong University School of MedicineShanghaiPR China
| | - Yaqiong Wang
- Department of Nephrology, Zhongshan HospitalFudan UniversityShanghaiPR China
| | - Zhaoyang Chen
- Cardiology department, Union HospitalFujian Medical UniversityFuzhouPR China
| | - Zhigang Li
- Department of Cardiovascular Medicine, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiPR China,Institute of Cardiovascular DiseasesShanghai Jiao Tong University School of MedicineShanghaiPR China
| | - Ziyang Wang
- Department of Cardiovascular Medicine, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiPR China,Institute of Cardiovascular DiseasesShanghai Jiao Tong University School of MedicineShanghaiPR China
| | - Kangni Jia
- Department of Cardiovascular Medicine, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiPR China,Institute of Cardiovascular DiseasesShanghai Jiao Tong University School of MedicineShanghaiPR China
| | - Jiaxin Zhao
- Department of Cardiovascular Medicine, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiPR China,Institute of Cardiovascular DiseasesShanghai Jiao Tong University School of MedicineShanghaiPR China
| | - Hang Zhang
- Department of Cardiovascular Medicine, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiPR China,Institute of Cardiovascular DiseasesShanghai Jiao Tong University School of MedicineShanghaiPR China
| | - Hongyang Xie
- Department of Cardiovascular Medicine, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiPR China,Institute of Cardiovascular DiseasesShanghai Jiao Tong University School of MedicineShanghaiPR China
| | - Lin Lu
- Department of Cardiovascular Medicine, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiPR China,Institute of Cardiovascular DiseasesShanghai Jiao Tong University School of MedicineShanghaiPR China
| | - Kang Chen
- Department of Cardiovascular Medicine, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiPR China
| | - Lei Chen
- Shanghai Institute of ImmunologyShanghai Jiao Tong University School of MedicineShanghaiPR China
| | - Keiichi Fukuda
- Department of CardiologyKeio University School of MedicineTokyoJapan
| | - Motoaki Sano
- Department of CardiologyKeio University School of MedicineTokyoJapan
| | - Ruiyan Zhang
- Department of Cardiovascular Medicine, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiPR China,Institute of Cardiovascular DiseasesShanghai Jiao Tong University School of MedicineShanghaiPR China
| | - Jun Liu
- Department of Cardiovascular Surgery, Shanghai East HospitalTongji University School of MedicineShanghaiPR China
| | - Xiaoxiang Yan
- Department of Cardiovascular Medicine, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiPR China,Institute of Cardiovascular DiseasesShanghai Jiao Tong University School of MedicineShanghaiPR China
| |
Collapse
|
11
|
Avery TY, Köhler N, Zeiser R, Brummer T, Ruess DA. Onco-immunomodulatory properties of pharmacological interference with RAS-RAF-MEK-ERK pathway hyperactivation. Front Oncol 2022; 12:931774. [PMID: 35965494 PMCID: PMC9363660 DOI: 10.3389/fonc.2022.931774] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/30/2022] [Indexed: 12/25/2022] Open
Abstract
Hyperactivation of the RAS-RAF-MEK-ERK cascade - a mitogen-activated protein kinase pathway – has a well-known association with oncogenesis of leading tumor entities, including non-small cell lung cancer, colorectal carcinoma, pancreatic ductal adenocarcinoma, and malignant melanoma. Increasing evidence shows that genetic alterations leading to RAS-RAF-MEK-ERK pathway hyperactivation mediate contact- and soluble-dependent crosstalk between tumor, tumor microenvironment (TME) and the immune system resulting in immune escape mechanisms and establishment of a tumor-sustaining environment. Consequently, pharmacological interruption of this pathway not only leads to tumor-cell intrinsic disruptive effects but also modification of the TME and anti-tumor immunomodulation. At the same time, the importance of ERK signaling in immune cell physiology and potentiation of anti-tumor immune responses through ERK signaling inhibition within immune cell subsets has received growing appreciation. Specifically, a strong case was made for targeted MEK inhibition due to promising associated immune cell intrinsic modulatory effects. However, the successful transition of therapeutic agents interrupting RAS-RAF-MEK-ERK hyperactivation is still being hampered by significant limitations regarding durable efficacy, therapy resistance and toxicity. We here collate and summarize the multifaceted role of RAS-RAF-MEK-ERK signaling in physiology and oncoimmunology and outline the rationale and concepts for exploitation of immunomodulatory properties of RAS-RAF-MEK-ERK inhibition while accentuating the role of MEK inhibition in combinatorial and intermittent anticancer therapy. Furthermore, we point out the extensive scientific efforts dedicated to overcoming the challenges encountered during the clinical transition of various therapeutic agents in the search for the most effective and safe patient- and tumor-tailored treatment approach.
Collapse
Affiliation(s)
- Thomas Yul Avery
- Department of General and Visceral Surgery, Center of Surgery, Medical Center University of Freiburg, Freiburg, Germany
- *Correspondence: Thomas Yul Avery, ; Dietrich Alexander Ruess,
| | - Natalie Köhler
- Department of Medicine I - Medical Center, Medical Center University of Freiburg, Freiburg, Germany
- CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Robert Zeiser
- Department of Medicine I - Medical Center, Medical Center University of Freiburg, Freiburg, Germany
- German Cancer Consortium Deutsches Konsortium Translationale Krebsforschung (DKTK), partner site Freiburg, German Cancer Research Center Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Tilman Brummer
- German Cancer Consortium Deutsches Konsortium Translationale Krebsforschung (DKTK), partner site Freiburg, German Cancer Research Center Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
- Institute of Molecular Medicine and Cell Research (IMMZ), Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Comprehensive Cancer Center Freiburg (CCCF), Faculty of Medicine, Medical Center University of Freiburg, Freiburg, Germany
| | - Dietrich Alexander Ruess
- Department of General and Visceral Surgery, Center of Surgery, Medical Center University of Freiburg, Freiburg, Germany
- German Cancer Consortium Deutsches Konsortium Translationale Krebsforschung (DKTK), partner site Freiburg, German Cancer Research Center Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
- *Correspondence: Thomas Yul Avery, ; Dietrich Alexander Ruess,
| |
Collapse
|
12
|
Gama-Cuellar AG, Francisco ALN, Scarini JF, Mariano FV, Kowalski LP, Gondak R. Decreased CD1a + and CD83 + cells in tonsillar squamous cell carcinoma regardless of HPV status. J Appl Oral Sci 2022; 30:e20210702. [PMID: 35584505 PMCID: PMC9126112 DOI: 10.1590/1678-7757-2020-0702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 04/06/2022] [Indexed: 11/22/2022] Open
Abstract
Dendritic cells (DCs) are specialized antigen-presenting cells that play a critical role in the immune response against human papillomavirus (HPV) infection, and represent a therapeutic target in cancer.
Collapse
Affiliation(s)
| | | | - João Figueira Scarini
- Universidade Estadual de Campinas - UNICAMP, Departamento de Anatomia-Patológica, Faculdade de Ciências Médicas, Campinas, SP, Brasil
| | - Fernanda Viviane Mariano
- Universidade Estadual de Campinas - UNICAMP, Departamento de Anatomia-Patológica, Faculdade de Ciências Médicas, Campinas, SP, Brasil
| | | | - Rogério Gondak
- Universidade Federal de Santa Catarina - UFSC, Departamento de Patologia, Florianópolis, SC, Brasil
| |
Collapse
|
13
|
Dendritic Cells and Their Immunotherapeutic Potential for Treating Type 1 Diabetes. Int J Mol Sci 2022; 23:ijms23094885. [PMID: 35563276 PMCID: PMC9099521 DOI: 10.3390/ijms23094885] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/22/2022] [Accepted: 04/26/2022] [Indexed: 12/15/2022] Open
Abstract
Type 1 diabetes (T1D) results from the destruction of pancreatic beta cells through a process that is primarily mediated by T cells. Emerging evidence suggests that dendritic cells (DCs) play a crucial role in initiating and developing this debilitating disease. DCs are professional antigen-presenting cells with the ability to integrate signals arising from tissue infection or injury that present processed antigens from these sites to naïve T cells in secondary lymphoid organs, thereby triggering naïve T cells to differentiate and modulate adaptive immune responses. Recent advancements in our knowledge of the various subsets of DCs and their cellular structures and methods of orchestration over time have resulted in a better understanding of how the T cell response is shaped. DCs employ various arsenal to maintain their tolerance, including the induction of effector T cell deletion or unresponsiveness and the generation and expansion of regulatory T cell populations. Therapies that suppress the immunogenic effects of dendritic cells by blocking T cell costimulatory pathways and proinflammatory cytokine production are currently being sought. Moreover, new strategies are being developed that can regulate DC differentiation and development and harness the tolerogenic capacity of these cells. Here, in this report, we focus on recent advances in the field of DC immunology and evaluate the prospects of DC-based therapeutic strategies to treat T1D.
Collapse
|
14
|
The Influence of Antibiotic Resistance on Innate Immune Responses to Staphylococcus aureus Infection. Antibiotics (Basel) 2022; 11:antibiotics11050542. [PMID: 35625186 PMCID: PMC9138074 DOI: 10.3390/antibiotics11050542] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/13/2022] [Accepted: 04/18/2022] [Indexed: 11/16/2022] Open
Abstract
Staphylococcus aureus (S. aureus) causes a broad range of infections and is associated with significant morbidity and mortality. S. aureus produces a diverse range of cellular and extracellular factors responsible for its invasiveness and ability to resist immune attack. In recent years, increasing resistance to last-line anti-staphylococcal antibiotics daptomycin and vancomycin has been observed. Resistant strains of S. aureus are highly efficient in invading a variety of professional and nonprofessional phagocytes and are able to survive inside host cells. Eliciting immune protection against antibiotic-resistant S. aureus infection is a global challenge, requiring both innate and adaptive immune effector mechanisms. Dendritic cells (DC), which sit at the interface between innate and adaptive immune responses, are central to the induction of immune protection against S. aureus. However, it has been observed that S. aureus has the capacity to develop further antibiotic resistance and acquire increased resistance to immunological recognition by the innate immune system. In this article, we review the strategies utilised by S. aureus to circumvent antibiotic and innate immune responses, especially the interaction between S. aureus and DC, focusing on how this relationship is perturbed with the development of antibiotic resistance.
Collapse
|
15
|
Khilji MS, Faridi P, Pinheiro-Machado E, Hoefner C, Dahlby T, Aranha R, Buus S, Nielsen M, Klusek J, Mandrup-Poulsen T, Pandey K, Purcell AW, Marzec MT. Defective Proinsulin Handling Modulates the MHC I Bound Peptidome and Activates the Inflammasome in β-Cells. Biomedicines 2022; 10:biomedicines10040814. [PMID: 35453564 PMCID: PMC9024965 DOI: 10.3390/biomedicines10040814] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/23/2022] [Accepted: 03/28/2022] [Indexed: 12/04/2022] Open
Abstract
How immune tolerance is lost to pancreatic β-cell peptides triggering autoimmune type 1 diabetes is enigmatic. We have shown that loss of the proinsulin chaperone glucose-regulated protein (GRP) 94 from the endoplasmic reticulum (ER) leads to mishandling of proinsulin, ER stress, and activation of the immunoproteasome. We hypothesize that inadequate ER proinsulin folding capacity relative to biosynthetic need may lead to an altered β-cell major histocompatibility complex (MHC) class-I bound peptidome and inflammasome activation, sensitizing β-cells to immune attack. We used INS-1E cells with or without GRP94 knockout (KO), or in the presence or absence of GRP94 inhibitor PU-WS13 (GRP94i, 20 µM), or exposed to proinflammatory cytokines interleukin (IL)-1β or interferon gamma (IFNγ) (15 pg/mL and 10 ng/mL, respectively) for 24 h. RT1.A (rat MHC I) expression was evaluated using flow cytometry. The total RT1.A-bound peptidome analysis was performed on cell lysates fractionated by reverse-phase high-performance liquid chromatography (RP-HPLC), followed by liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS). The nucleotide-binding oligomerization domain, leucine rich repeat and pyrin domain containing protein (NLRP1), nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor alpha (IκBα), and (pro) IL-1β expression and secretion were investigated by Western blotting. GRP94 KO increased RT1.A expression in β-cells, as did cytokine exposure compared to relevant controls. Immunopeptidome analysis showed increased RT1.A-bound peptide repertoire in GRP94 KO/i cells as well as in the cells exposed to cytokines. The GRP94 KO/cytokine exposure groups showed partial overlap in their peptide repertoire. Notably, proinsulin-derived peptide diversity increased among the total RT1.A peptidome in GRP94 KO/i along with cytokines exposure. NLRP1 expression was upregulated in GRP94 deficient cells along with decreased IκBα content while proIL-1β cellular levels declined, coupled with increased secretion of mature IL-1β. Our results suggest that limiting β-cell proinsulin chaperoning enhances RT1.A expression alters the MHC-I peptidome including proinsulin peptides and activates inflammatory pathways, suggesting that stress associated with impeding proinsulin handling may sensitize β-cells to immune-attack.
Collapse
Affiliation(s)
- Muhammad Saad Khilji
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (M.S.K.); (C.H.); (T.M.-P.)
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3168, Australia; (R.A.); (K.P.)
- Department of Physiology, University of Veterinary and Animal Sciences, Lahore 54000, Pakistan
| | - Pouya Faridi
- Department of Medicine, School of Clinical Sciences, Monash Univesity, Clayton, VIC 3168, Australia;
| | - Erika Pinheiro-Machado
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, 9713 Groningen, The Netherlands;
| | - Carolin Hoefner
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (M.S.K.); (C.H.); (T.M.-P.)
| | - Tina Dahlby
- Laboratory of Translational Nutrition Biology, Department of Health Sciences and Technology, Institute of Food, Nutrition and Health, ETH Zürich, 8603 Zürich, Switzerland;
| | - Ritchlynn Aranha
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3168, Australia; (R.A.); (K.P.)
| | - Søren Buus
- Department of Immunology and Microbiology, University of Copenhagen, 2200 Copenhagen, Denmark;
| | - Morten Nielsen
- Department of Health Technology, Section for Bioinformatics, Technical University of Denmark, 2800 Lyngby, Denmark;
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín, San Martín CP1650, Argentina
| | - Justyna Klusek
- Laboratory of Medical Genetics, Department of Surgical Medicine, Collegium Medicum, Jan Kochanowski University, 25-369 Kielce, Poland;
| | - Thomas Mandrup-Poulsen
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (M.S.K.); (C.H.); (T.M.-P.)
| | - Kirti Pandey
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3168, Australia; (R.A.); (K.P.)
| | - Anthony W. Purcell
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3168, Australia; (R.A.); (K.P.)
- Correspondence: (A.W.P.); (M.T.M.); Tel.: +61-39-902-9265 (A.W.P.); +45-25-520-256 (M.T.M.)
| | - Michal T. Marzec
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (M.S.K.); (C.H.); (T.M.-P.)
- Institute of Health Sciences, Collegium Medicum, Jan Kochanowski University, 25-002 Kielce, Poland
- Correspondence: (A.W.P.); (M.T.M.); Tel.: +61-39-902-9265 (A.W.P.); +45-25-520-256 (M.T.M.)
| |
Collapse
|
16
|
Xu Y, Ma S, Zhao J, Chen H, Si X, Huang Z, Yu Z, Song W, Tang Z, Chen X. Mannan-decorated pathogen-like polymeric nanoparticles as nanovaccine carriers for eliciting superior anticancer immunity. Biomaterials 2022; 284:121489. [PMID: 35364489 DOI: 10.1016/j.biomaterials.2022.121489] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 03/20/2022] [Accepted: 03/25/2022] [Indexed: 12/20/2022]
Abstract
Using nanotechnology for cancer vaccine design holds great promise because of the intrinsic feature of nanoparticles in being captured by antigen-presenting cells (APCs). However, there are still obstacles in current nanovaccine systems in achieving efficient tumor therapeutic effects, which could partially be attributed to the unsatisfactory vaccine carrier design. Herein, we report a mannan-decorated pathogen-like polymeric nanoparticle as a protein vaccine carrier for eliciting robust anticancer immunity. This nanovaccine was constructed as a core-shell structure with mannan as the shell, polylactic acid-polyethylenimine (PLA-PEI) assembled nanoparticle as the core, and protein antigens and Toll-like receptor 9 (TLR9) agonist CpG absorbed onto the PLA-PEI core via electrostatic interactions. Compared to other hydrophilic materials, mannan decoration could greatly enhance the lymph node draining ability of the nanovaccine and promote the capturing by the CD8+ dendritic cells (DCs) in the lymph node, while PLA-PEI as the inner core could enhance antigen endosome escape thus promoting the antigen cross-presentation. In addition, mannan itself as a TLR4 agonist could synergize with CpG for maximally activating the DCs. Excitingly, we observed in several murine tumor models that using this nanovaccine alone could elicit robust immune response in vivo and result in superior anti-tumor effects with 50% of mice completely cured. This study strongly evidenced that mannan decoration and a rationally designed nanovaccine system could be quite robust in tumor vaccine therapy.
Collapse
Affiliation(s)
- Yudi Xu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China; University of Chinese Academy of Sciences, Beijing, 100039, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, China
| | - Sheng Ma
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, China
| | - Jiayu Zhao
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China; University of Science and Technology of China, Hefei, 230026, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, China
| | - Hongyu Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China; University of Science and Technology of China, Hefei, 230026, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, China
| | - Xinghui Si
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China; University of Science and Technology of China, Hefei, 230026, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, China
| | - Zichao Huang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China; University of Science and Technology of China, Hefei, 230026, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, China
| | - Zhentao Yu
- Department of Gastrointestinal and Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Wantong Song
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, China.
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China; University of Science and Technology of China, Hefei, 230026, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China; University of Science and Technology of China, Hefei, 230026, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, China
| |
Collapse
|
17
|
He Y, Hu X, Zhang H, Chen X, Sun H. Adjuvant effect of two polysaccharides from the petals of Crocus sativus and its mechanisms. Int J Biol Macromol 2022; 204:50-61. [PMID: 35122804 DOI: 10.1016/j.ijbiomac.2022.01.169] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 01/22/2022] [Accepted: 01/28/2022] [Indexed: 12/21/2022]
Abstract
Two polysaccharides from Crocus sativus petals (PCSPs), PCSPA and PCSPB, presented immunopotentiatory activity through activating macrophages via MAPK and NF-κB pathway. In this study, two PCSPs were investigated for the adjuvant effects and underlying mechanisms using ovalbumin (OVA) in mice. PCSPA and PCSPB remarkably not only boosted the OVA-specific IgG antibody and its isotype titers, but strengthened splenocyte proliferation and natural killer cell activities in immunized mice. Both PCSPs also dramatically triggered the production of Th1- and Th2-cytokines and facilitated the gene expression of Th1- and Th2-cytokines and transcription factors in OVA-stimulated splenocytes. In mechanisms, two PCSPs rapidly elicited the gene and protein expression of cytokines and chemokines, promoted the recruitment and antigen uptake of immune cells in the injected-muscles, and augmented the migration and antigen transport of immune cells to the draining lymph nodes. These findings demonstrated that PCSPs enhanced and improved immune responses and simultaneously elicited Th1- and Th2-immune responses to OVA through activating innate immune microenvironment, and that they could act as promising vaccine adjuvant candidates.
Collapse
Affiliation(s)
- Yanfei He
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; College of Biotechnology and Pharmaceutical Engineering, West Anhui University, Lu'an 237012, China
| | - Xiaoying Hu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Huifang Zhang
- Medical College, Jinhua Polytechnic, Jinhua 321000, China
| | - Xiangfeng Chen
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Hongxiang Sun
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
18
|
Li R, Hu X, Chen H, Zhao Y, Gao X, Yuan Y, Guo H, Huang H, Zou X, Qi H, Liu H, Shang Y. Role of Cholinergic Anti-Inflammatory Pathway in Protecting Sepsis-Induced Acute Lung Injury through Regulation of the Conventional Dendritic Cells. Mediators Inflamm 2022; 2022:1474891. [PMID: 35125962 PMCID: PMC8813293 DOI: 10.1155/2022/1474891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 01/03/2022] [Accepted: 01/06/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND The cholinergic anti-inflammatory pathway connects the immune response system and the nervous system via the vagus nerve. The key regulatory receptor is the α7-subtype of the nicotinic acetylcholine receptor (α7nAChR). Cholinergic anti-inflammatory pathway has been proved to be effective in suppressing the inflammation responses in acute lung injury (ALI). Dendritic cells (DCs), the important antigen-presenting cells, also express the α7nAChR. Past studies have indicated that reducing the quantity of mature conventional DCs and inhibiting the maturation of pulmonary DCs may prove effective for the treatment of ALI. However, the effects of cholinergic anti-inflammatory pathway on maturation, function, and quantity of DCs and conventional DCs in ALI remain unclear. OBJECTIVE It was hypothesized that cholinergic anti-inflammatory pathway may inhibit the inflammatory response of ALI by regulating maturation, phenotype, and quantity of DCs and conventional DCs. METHODS GTS-21 (GTS-21 dihydrochloride), an α7nAchR agonist, was prophylactically administered in sepsis-induced ALI mouse model and LPS-primed bone marrow-derived dendritic cells. The effects of GTS-21 were observed with respect to maturation, phenotype, and quantity of DCs, conventional DCs, and conventional DCs2 (type 2 conventional DCs) and the release of DC-related proinflammatory cytokines in vivo and in vitro. RESULTS The results of the present study revealed that GTS-21 treatment decreased the maturation of DCs and the production of DC-related proinflammatory cytokines in vitro and in sepsis-induced ALI mouse model; it reduced the quantity of CD11c+MHCII+ conventional DCs and CD11c+CD11b+ conventional DCs2 in vivo experiment. CONCLUSIONS Cholinergic anti-inflammatory pathway contributes to the reduction in the inflammatory response in ALI by regulating maturation, phenotype, and quantity of DCs, conventional DCs, and conventional DCs2.
Collapse
Affiliation(s)
- Ruiting Li
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Xuemei Hu
- Department of Nephrology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province 442000, China
| | - Huibin Chen
- Department of Critical Care Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province 442000, China
| | - Yue Zhao
- Department of Critical Care Medicine, Jin Yin-tan Hospital, Wuhan, Hubei 430048, China
| | - Xuehui Gao
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Yin Yuan
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Huiling Guo
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Haiyan Huang
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Xiaojing Zou
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Hong Qi
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Hong Liu
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - You Shang
- Department of Critical Care Medicine, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| |
Collapse
|
19
|
Neuropilin-1, a myeloid cell-specific protein, is an inhibitor of HIV-1 infectivity. Proc Natl Acad Sci U S A 2022; 119:2114884119. [PMID: 34987100 PMCID: PMC8764665 DOI: 10.1073/pnas.2114884119] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2021] [Indexed: 12/13/2022] Open
Abstract
Myeloid lineage cells such as macrophages and dendritic cells (DCs), targeted by HIV-1, are important vehicles for virus dissemination through the body as well as viral reservoirs. Compared to activated lymphocytes, myeloid cells are collectively more resistant to HIV-1 infection. Here we report that NRP-1, encoding transmembrane protein neuropilin-1, is highly expressed in macrophages and DCs but not CD4+ T cells, serving as an anti-HIV factor to inhibit the infectivity of HIV-1 progeny virions. Silencing NRP-1 enhanced the transmission of HIV-1 in macrophages and DCs significantly and increased the infectivity of the virions produced by these cells. We further demonstrated that NRP-1 was packaged into the progeny virions to inhibit their ability to attach to target cells, thus reducing the infectivity of the virions. These data indicate that NRP-1 is a newly identified antiviral protein highly produced in both macrophages and DCs that inhibit HIV-1 infectivity; thus, NRP-1 may be a novel therapeutic strategy for the treatment of HIV-1 infection.
Collapse
|
20
|
Arabpour M, Lebrero-Fernandez C, Schön K, Strömberg A, Börjesson V, Lahl K, Ballegeer M, Saelens X, Angeletti D, Agace W, Lycke N. ADP-ribosylating adjuvant reveals plasticity in cDC1 cells that drive mucosal Th17 cell development and protection against influenza virus infection. Mucosal Immunol 2022; 15:745-761. [PMID: 35418673 PMCID: PMC9259495 DOI: 10.1038/s41385-022-00510-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 03/07/2022] [Accepted: 03/23/2022] [Indexed: 02/04/2023]
Abstract
Migratory dendritic cells expressing CD103 are the targets for mucosal vaccines. These belong to either of two lineage-restricted subsets, cDC1 or cDC2 cells, which have been linked to priming of functionally distinct CD4 T cells. However, recent studies have identified plasticity in cDC2 cells with overlapping functions with cDC1 cells, while the converse has not been reported. We genetically engineered a vaccine adjuvant platform that targeted the cholera toxin A1 (CTA1) ADP-ribosylating enzyme to CD103+ cDC1 and cDC2 cells using a single-chain antibody (scFv) to CD103. Unexpectedly, intranasal immunization with the CTA1-svFcCD103 adjuvant modified cDC1 cells to effectively prime Th17 cells, a function previously limited to cDC2 cells. In fact, cDC2 cells were dispensible, while cDC1 cells, lacking in Batf3-/- mice, were critical. Following intranasal immunizations isolated cDC1 cells from mLN exclusively promoted Rorgt+ T cells and IL-17, IL-21, and IL-22 production. Strong CD8 T cell responses through antigen cross presentation by cDC1 cells were also observed. Single-cell RNAseq analysis revealed upregulation of Th17-promoting gene signatures in sorted cDC1 cells. Gene expression in isolated cDC2 cells was largely unaffected. Our finding represents a major shift of paradigm as we have documented functional plasticity in cDC1 cells.
Collapse
Affiliation(s)
- Mohammad Arabpour
- grid.8761.80000 0000 9919 9582MIVAC-Mucosal Immunobiology & Vaccine Center, Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Cristina Lebrero-Fernandez
- grid.8761.80000 0000 9919 9582MIVAC-Mucosal Immunobiology & Vaccine Center, Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Karin Schön
- grid.8761.80000 0000 9919 9582MIVAC-Mucosal Immunobiology & Vaccine Center, Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Anneli Strömberg
- grid.8761.80000 0000 9919 9582MIVAC-Mucosal Immunobiology & Vaccine Center, Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Vanja Börjesson
- grid.8761.80000 0000 9919 9582Bioinformatics Core Facility, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Katharina Lahl
- grid.4514.40000 0001 0930 2361Immunology Section, Lund University, BMC D14, 221-84 Lund, Sweden
| | - Marlies Ballegeer
- grid.5342.00000 0001 2069 7798VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium and Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Xavier Saelens
- grid.5342.00000 0001 2069 7798VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium and Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Davide Angeletti
- grid.8761.80000 0000 9919 9582MIVAC-Mucosal Immunobiology & Vaccine Center, Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - William Agace
- grid.4514.40000 0001 0930 2361Immunology Section, Lund University, BMC D14, 221-84 Lund, Sweden ,grid.5170.30000 0001 2181 8870Mucosal Immunology Group, Department of Health Technology, Technical University of Denmark, Kemitorvet, 2800 Kgs, Lyngby, Denmark
| | - Nils Lycke
- grid.8761.80000 0000 9919 9582MIVAC-Mucosal Immunobiology & Vaccine Center, Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
21
|
Guo B, Peng Y, Gu Y, Zhong Y, Su C, Liu L, Chai D, Song T, Zhao N, Yan X, Xu T. Resveratrol pretreatment mitigates LPS-induced acute lung injury by regulating conventional dendritic cells' maturation and function. Open Life Sci 2021; 16:1064-1081. [PMID: 34676301 PMCID: PMC8483064 DOI: 10.1515/biol-2021-0110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/10/2021] [Accepted: 08/16/2021] [Indexed: 11/20/2022] Open
Abstract
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is a severe syndrome lacking efficient therapy and resulting in high morbidity and mortality. Although resveratrol (RES), a natural phytoalexin, has been reported to protect the ALI by suppressing the inflammatory response, the detailed mechanism of how RES affected the immune system is poorly studied. Pulmonary conventional dendritic cells (cDCs) are critically involved in the pathogenesis of inflammatory lung diseases including ALI. In this study, we aimed to investigate the protective role of RES via pulmonary cDCs in lipopolysaccharide (LPS)-induced ALI mice. Murine ALI model was established by intratracheally challenging with 5 mg/kg LPS. We found that RES pretreatment could mitigate LPS-induced ALI. Additionally, proinflammatory-skewed cytokines decreased whereas anti-inflammatory-related cytokines increased in bronchoalveolar lavage fluid by RES pretreatment. Mechanistically, RES regulated pulmonary cDCs' maturation and function, exhibiting lower level of CD80, CD86, major histocompatibility complex (MHC) II expression, and IL-10 secretion in ALI mice. Furthermore, RES modulated the balance between proinflammation and anti-inflammation of cDCs. Moreover, in vitro RES pretreatment regulated the maturation and function of bone marrow derived dendritic cells (BMDCs). Finally, the adoptive transfer of RES-pretreated BMDCs enhanced recovery of ALI. Thus, these data might further extend our understanding of a protective role of RES in regulating pulmonary cDCs against ALI.
Collapse
Affiliation(s)
- Bingnan Guo
- Jiangsu Institute of Health Emergency, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
- Department of Emergency Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Yigen Peng
- Department of Emergency Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu 211100, China
| | - Yuting Gu
- Jiangsu Institute of Health Emergency, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
- Department of Emergency Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Yi Zhong
- Jiangsu Institute of Health Emergency, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
- Department of Emergency Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Chenglei Su
- Jiangsu Institute of Health Emergency, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
- Department of Emergency Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Lin Liu
- Jiangsu Institute of Health Emergency, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
- Department of Emergency Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Dafei Chai
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Tengfei Song
- The Feinstein Institute for Medical Research, Manhasset, NY 11030, New York, United States
| | - Ningjun Zhao
- Jiangsu Institute of Health Emergency, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
- Department of Emergency Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Xianliang Yan
- Jiangsu Institute of Health Emergency, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
- Department of Emergency Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Tie Xu
- Jiangsu Institute of Health Emergency, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
- Department of Emergency Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
- Department of Emergency Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu 211100, China
| |
Collapse
|
22
|
Pecora F, Abate L, Scavone S, Petrucci I, Costa F, Caminiti C, Argentiero A, Esposito S. Management of Infectious Lymphadenitis in Children. CHILDREN-BASEL 2021; 8:children8100860. [PMID: 34682125 PMCID: PMC8535130 DOI: 10.3390/children8100860] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/22/2021] [Accepted: 09/22/2021] [Indexed: 01/22/2023]
Abstract
Lymphadenopathy is an irregularity in the size and texture of the lymph nodes, which is quite common in childhood. When the enlargement of lymph nodes is caused by inflammatory and infectious processes, it is called lymphadenitis. The main objective of this manuscript is to summarize the common infectious etiologies and presentations of lymphadenitis in children providing a management guide for clinical practice. PubMed was used to search for all of the studies published up to April 2021 using keywords such as "lymphadenitis" and "children". Literature analysis showed that the differential diagnosis for lymphadenitis in pediatrics is broad. Although lymph node enlargement in children is usually benign and self-limited, it is important to exclude malignant etiology. In most cases, history and physical examination allow to identify the correct diagnosis and start a proper treatment with a prompt resolution of the lymphadenopathy. However, particularly in the case of persistent lymphadenitis, determining the cause of lymph node enlargement may be difficult, and the exact etiology may not be identified despite extensive investigations. Further studies should develop and validate an algorithm to assist pediatricians in the diagnosis and timely treatment of lymphadenitis, suggesting situations in which a watchful waiting may be considered a safe approach, those in which empiric antibiotic therapy should be administered, and those requiring a timely diagnostic work-up.
Collapse
Affiliation(s)
- Francesco Pecora
- Paediatric Clinic, Pietro Barilla Children’s Hospital, Department of Medicine and Surgery, University of Parma, via Gramsci 14, 43126 Parma, Italy; (F.P.); (L.A.); (S.S.); (I.P.); (F.C.); (A.A.)
| | - Luciana Abate
- Paediatric Clinic, Pietro Barilla Children’s Hospital, Department of Medicine and Surgery, University of Parma, via Gramsci 14, 43126 Parma, Italy; (F.P.); (L.A.); (S.S.); (I.P.); (F.C.); (A.A.)
| | - Sara Scavone
- Paediatric Clinic, Pietro Barilla Children’s Hospital, Department of Medicine and Surgery, University of Parma, via Gramsci 14, 43126 Parma, Italy; (F.P.); (L.A.); (S.S.); (I.P.); (F.C.); (A.A.)
| | - Irene Petrucci
- Paediatric Clinic, Pietro Barilla Children’s Hospital, Department of Medicine and Surgery, University of Parma, via Gramsci 14, 43126 Parma, Italy; (F.P.); (L.A.); (S.S.); (I.P.); (F.C.); (A.A.)
| | - Federico Costa
- Paediatric Clinic, Pietro Barilla Children’s Hospital, Department of Medicine and Surgery, University of Parma, via Gramsci 14, 43126 Parma, Italy; (F.P.); (L.A.); (S.S.); (I.P.); (F.C.); (A.A.)
| | - Caterina Caminiti
- Research an Innovation Unit, University Hospital of Parma, via Gramsci 14, 43126 Parma, Italy;
| | - Alberto Argentiero
- Paediatric Clinic, Pietro Barilla Children’s Hospital, Department of Medicine and Surgery, University of Parma, via Gramsci 14, 43126 Parma, Italy; (F.P.); (L.A.); (S.S.); (I.P.); (F.C.); (A.A.)
| | - Susanna Esposito
- Paediatric Clinic, Pietro Barilla Children’s Hospital, Department of Medicine and Surgery, University of Parma, via Gramsci 14, 43126 Parma, Italy; (F.P.); (L.A.); (S.S.); (I.P.); (F.C.); (A.A.)
- Correspondence: ; Tel.: +39-0521-903-524
| |
Collapse
|
23
|
Plantinga M, van den Beemt DAMH, Dünnebach E, Nierkens S. CD14 Expressing Precursors Give Rise to Highly Functional Conventional Dendritic Cells for Use as Dendritic Cell Vaccine. Cancers (Basel) 2021; 13:cancers13153818. [PMID: 34359719 PMCID: PMC8345076 DOI: 10.3390/cancers13153818] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 01/29/2023] Open
Abstract
Simple Summary Dendritic cells are attractive candidates for immunotherapy to prevent disease recurrence in cancer patients. Dendritic cells are a plastic population of antigen presenting cells and a variety of protocols have been described to generate dendritic cells from either monocytes or stem cells. To induce long lasting immunity, dendritic cells need to have a fully mature phenotype and activate naïve T-cells. Here, we describe a good manufacturer protocol to generate very potent conventional DC-like cells, derived from cord blood stem cells via a CD14+CD115+ precursor stage. They express high levels of CD1c and strongly activate both naïve as well as antigen-experienced T-cells. Implementation of this protocol in the clinic could advance the efficiency of dendritic cell based vaccines. Abstract Induction of long-lasting immunity by dendritic cells (DCs) makes them attractive candidates for anti-tumor vaccination. Although DC vaccinations are generally considered safe, clinical responses remain inconsistent in clinical trials. This initiated studies to identify subsets of DCs with superior capabilities to induce effective and memory anti-tumor responses. The use of primary DCs has been suggested to overcome the functional limitations of ex vivo monocyte-derived DCs (moDC). The ontogeny of primary DCs has recently been revised by the introduction of DC3, which phenotypically resembles conventional (c)DC2 as well as moDC. Previously, we developed a protocol to generate cDC2s from cord blood (CB)-derived stem cells via a CD115-expressing precursor. Here, we performed index sorting and single-cell RNA-sequencing to define the heterogeneity of in vitro developed DC precursors and identified CD14+CD115+ expressing cells that develop into CD1c++DCs and the remainder cells brought about CD123+DCs, as well as assessed their potency. The maturation status and T-cell activation potential were assessed using flow cytometry. CD123+DCs were specifically prone to take up antigens but only modestly activated T-cells. In contrast, CD1c++ are highly mature and specialized in both naïve as well as antigen-experienced T-cell activation. These findings show in vitro functional diversity between cord blood stem cell-derived CD123+DC and CD1c++DCs and may advance the efficiency of DC-based vaccines.
Collapse
Affiliation(s)
- Maud Plantinga
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
| | - Denise A M H van den Beemt
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
| | - Ester Dünnebach
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
| | - Stefan Nierkens
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands
| |
Collapse
|
24
|
Role of Dendritic Cell in Diabetic Nephropathy. Int J Mol Sci 2021; 22:ijms22147554. [PMID: 34299173 PMCID: PMC8308035 DOI: 10.3390/ijms22147554] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/09/2021] [Accepted: 07/10/2021] [Indexed: 11/18/2022] Open
Abstract
Diabetic nephropathy (DN) is one of the most significant microvascular complications in diabetic patients. DN is the leading cause of end-stage renal disease, accounting for approximately 50% of incident cases. The current treatment options, such as optimal control of hyperglycemia and elevated blood pressure, are insufficient to prevent its progression. DN has been considered as a nonimmune, metabolic, or hemodynamic glomerular disease initiated by hyperglycemia. However, recent studies suggest that DN is an inflammatory disease, and immune cells related with innate and adaptive immunity, such as macrophage and T cells, might be involved in its development and progression. Although it has been revealed that kidney dendritic cells (DCs) accumulation in the renal tissue of human and animal models of DN require activated T cells in the kidney disease, little is known about the function of DCs in DN. In this review, we describe kidney DCs and their subsets, and the role in the pathogenesis of DN. We also suggest how to improve the kidney outcomes by modulating kidney DCs optimally in the patients with DN.
Collapse
|
25
|
Li N, Steiger S, Fei L, Li C, Shi C, Salei N, Schraml BU, Zheng Z, Anders HJ, Lichtnekert J. IRF8-Dependent Type I Conventional Dendritic Cells (cDC1s) Control Post-Ischemic Inflammation and Mildly Protect Against Post-Ischemic Acute Kidney Injury and Disease. Front Immunol 2021; 12:685559. [PMID: 34234783 PMCID: PMC8255684 DOI: 10.3389/fimmu.2021.685559] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 05/26/2021] [Indexed: 11/13/2022] Open
Abstract
Post-ischemic acute kidney injury and disease (AKI/AKD) involve acute tubular necrosis and irreversible nephron loss. Mononuclear phagocytes including conventional dendritic cells (cDCs) are present during different phases of injury and repair, but the functional contribution of this subset remains controversial. Transcription factor interferon regulatory factor 8 (IRF8) is required for the development of type I conventional dendritic cells (cDC1s) lineage and helps to define distinct cDC1 subsets. We identified one distinct subset among mononuclear phagocyte subsets according to the expression patterns of CD11b and CD11c in healthy kidney and lymphoid organs, of which IRF8 was significantly expressed in the CD11blowCD11chigh subset that mainly comprised cDC1s. Next, we applied a Irf8-deficient mouse line (Irf8fl/flClec9acre mice) to specifically target Clec9a-expressing cDC1s in vivo. During post-ischemic AKI/AKD, these mice lacked cDC1s in the kidney without affecting cDC2s. The absence of cDC1s mildly aggravated the loss of living primary tubule and decline of kidney function, which was associated with decreased anti-inflammatory Tregs-related immune responses, but increased T helper type 1 (TH1)-related and pro-inflammatory cytokines, infiltrating neutrophils and acute tubular cell death, while we also observed a reduced number of cytotoxic CD8+ T cells in the kidney when cDC1s were absent. Together, our data show that IRF8 is indispensable for kidney cDC1s. Kidney cDC1s mildly protect against post-ischemic AKI/AKD, probably via suppressing tissue inflammation and damage, which implies an immunoregulatory role for cDC1s.
Collapse
Affiliation(s)
- Na Li
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shen Zhen, China.,Division of Nephrology, Department of Medicine IV, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Stefanie Steiger
- Division of Nephrology, Department of Medicine IV, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Lingyan Fei
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shen Zhen, China
| | - Chenyu Li
- Division of Nephrology, Department of Medicine IV, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Chongxu Shi
- Division of Nephrology, Department of Medicine IV, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Natallia Salei
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany.,Institute for Cardiovascular Physiology and Pathophysiology, Biomedical Center, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Barbara U Schraml
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany.,Institute for Cardiovascular Physiology and Pathophysiology, Biomedical Center, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Zhihua Zheng
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shen Zhen, China
| | - Hans-Joachim Anders
- Division of Nephrology, Department of Medicine IV, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Julia Lichtnekert
- Division of Nephrology, Department of Medicine IV, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| |
Collapse
|
26
|
Bahadoran A, Bezavada L, Smallwood HS. Fueling influenza and the immune response: Implications for metabolic reprogramming during influenza infection and immunometabolism. Immunol Rev 2021; 295:140-166. [PMID: 32320072 DOI: 10.1111/imr.12851] [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: 12/31/2019] [Revised: 02/19/2020] [Accepted: 02/24/2020] [Indexed: 12/11/2022]
Abstract
Recent studies support the notion that glycolysis and oxidative phosphorylation are rheostats in immune cells whose bioenergetics have functional outputs in terms of their biology. Specific intrinsic and extrinsic molecular factors function as molecular potentiometers to adjust and control glycolytic to respiratory power output. In many cases, these potentiometers are used by influenza viruses and immune cells to support pathogenesis and the host immune response, respectively. Influenza virus infects the respiratory tract, providing a specific environmental niche, while immune cells encounter variable nutrient concentrations as they migrate in response to infection. Immune cell subsets have distinct metabolic programs that adjust to meet energetic and biosynthetic requirements to support effector functions, differentiation, and longevity in their ever-changing microenvironments. This review details how influenza coopts the host cell for metabolic reprogramming and describes the overlap of these regulatory controls in immune cells whose function and fate are dictated by metabolism. These details are contextualized with emerging evidence of the consequences of influenza-induced changes in metabolic homeostasis on disease progression.
Collapse
Affiliation(s)
- Azadeh Bahadoran
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Lavanya Bezavada
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Heather S Smallwood
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
27
|
Sharawi H, Heyman O, Mizraji G, Horev Y, Laviv A, Shapira L, Yona S, Hovav AH, Wilensky A. The Prevalence of Gingival Dendritic Cell Subsets in Periodontal Patients. J Dent Res 2021; 100:1330-1336. [PMID: 33899566 DOI: 10.1177/00220345211004864] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
As the most potent cells activating and polarizing naive T cells, dendritic cells (DCs) are of major importance in the induction of immunity and tolerance. DCs are a heterogeneous population of antigen-presenting cells that are widely distributed in lymphoid and nonlymphoid tissues. Murine studies have highlighted the important role of oral DCs and Langerhans cells (LCs) in orchestrating the physiological homeostasis of the oral mucosa. DCs are also critically involved in pathological conditions such as periodontal diseases, in which gingival DCs appear to have special localization and function. While the characterization of human DCs in health and disease has been extensively investigated in various tissues, this topic was rarely studied in human gingiva. Here, we employed an up-to-date approach to characterize by flow cytometry the gingival DCs of 27 healthy subjects and 21 periodontal patients. Four distinct subsets of mononuclear phagocytes were identified in healthy gingiva: conventional DC type 1 (cDC1), cDC2, plasmacytoid DCs (pDCs), and LCs. In periodontitis patients, the frequencies of gingival LCs and pDCs were dysregulated, as LCs decreased, whereas pDCs increased in the diseased gingiva. This shift in the prevalence of DCs was accompanied by increased expression of the proinflammatory cytokines interleukin (IL)-1β, interferon (IFN)-α, and IFN-γ, while the anti-inflammatory cytokine IL-10 was suppressed. We further found that smoking, a known risk factor of periodontitis, specifically reduces gingival LCs in healthy individuals, indicating a possible role of LCs in the elevated severity of periodontitis in smokers. Collectively, this work reveals the various DC subsets residing in the human gingiva and the impact of periodontitis, as well as smoking, on the prevalence of each subset. Our findings provide a foundation toward understanding the role of human DCs in orchestrating physiological oral immunity and set the stage for the evaluation and modulation of shifts in immunity associated with periodontitis.
Collapse
Affiliation(s)
- H Sharawi
- Department of Periodontology, The Hebrew University and Hadassah Medical Center, Jerusalem, Israel.,The Institute of Dental Sciences, The Hebrew University and Hadassah Medical Center, Jerusalem, Israel
| | - O Heyman
- Department of Periodontology, The Hebrew University and Hadassah Medical Center, Jerusalem, Israel.,The Institute of Dental Sciences, The Hebrew University and Hadassah Medical Center, Jerusalem, Israel
| | - G Mizraji
- Department of Periodontology, The Hebrew University and Hadassah Medical Center, Jerusalem, Israel.,The Institute of Dental Sciences, The Hebrew University and Hadassah Medical Center, Jerusalem, Israel
| | - Y Horev
- Department of Periodontology, The Hebrew University and Hadassah Medical Center, Jerusalem, Israel.,The Institute of Dental Sciences, The Hebrew University and Hadassah Medical Center, Jerusalem, Israel
| | - A Laviv
- Department of Oral and Maxillofacial Surgery, The Hebrew University and Hadassah Medical Center, Jerusalem, Israel.,The Department of Oral and Maxillofacial Surgery, The Maurice and Gabriela Goldschleger School of Dental Medicine, Tel Aviv University, Tel-Aviv, Israel
| | - L Shapira
- Department of Periodontology, The Hebrew University and Hadassah Medical Center, Jerusalem, Israel
| | - S Yona
- The Institute of Dental Sciences, The Hebrew University and Hadassah Medical Center, Jerusalem, Israel
| | - A H Hovav
- The Institute of Dental Sciences, The Hebrew University and Hadassah Medical Center, Jerusalem, Israel
| | - A Wilensky
- Department of Periodontology, The Hebrew University and Hadassah Medical Center, Jerusalem, Israel
| |
Collapse
|
28
|
Lin LCW, Croft SN, Croft NP, Wong YC, Smith SA, Tang SS, Purcell AW, Tscharke DC. Direct Priming of CD8 + T Cells Persists in the Face of Cowpox Virus Inhibitors of Antigen Presentation. J Virol 2021; 95:JVI.00186-21. [PMID: 33692206 PMCID: PMC8139650 DOI: 10.1128/jvi.00186-21] [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] [Received: 02/04/2021] [Accepted: 02/27/2021] [Indexed: 11/30/2022] Open
Abstract
Vaccinia virus (VACV) was the vaccine used to eradicate smallpox and is being repurposed as a vaccine vector. CD8+ T cells are key anti-viral mediators, but require priming to become effector or memory cells. Priming requires an interaction with dendritic cells that are either infected (direct priming), or that have acquired virus proteins but remain uninfected (cross priming). To investigate CD8+ T cell priming pathways for VACV, we engineered the virus to express CPXV12 and CPXV203, two inhibitors of antigen presentation encoded by cowpox virus. These intracellular proteins would be expected to block direct but not cross priming. The inhibitors had diverse impacts on the size of anti-VACV CD8+ T cell responses across epitopes and by different infection routes in mice, superficially suggesting variable use of direct and cross priming. However, when we then tested a form of antigen that requires direct priming, we found surprisingly that CD8+ T cell responses were not diminished by co-expression with CPXV12 and CPXV203. We then directly quantified the impact of CPXV12 and CPXV203 on viral antigen presentation using mass spectrometry, which revealed strong, but incomplete inhibition of antigen presentation by the CPXV proteins. Therefore, direct priming of CD8+ T cells by poxviruses is robust enough to withstand highly potent viral inhibitors of antigen presentation. This is a reminder of the limits of viral immune evasion and shows that viral inhibitors of antigen presentation cannot be assumed to dissect cleanly direct and cross priming of anti-viral CD8+ T cells.ImportanceCD8+ T cells are key to anti-viral immunity, so it is important to understand how they are activated. Many viruses have proteins that protect infected cells from T cell attack by interfering with the process that allows virus infection to be recognised by CD8+ T cells. It is thought that these proteins would also stop infected cells from activating T cells in the first place. However, we show here that this is not the case for two very powerful inhibitory proteins from cowpox virus. This demonstrates the flexibility and robustness of immune processes that turn on the immune responses required to fight infection.
Collapse
Affiliation(s)
- Leon C. W. Lin
- John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Sarah N. Croft
- John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Nathan P. Croft
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Yik Chun Wong
- John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Stewart A. Smith
- John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Swee-Seong Tang
- John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Anthony W. Purcell
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - David C. Tscharke
- John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| |
Collapse
|
29
|
Hathaway-Schrader JD, Novince CM. Maintaining homeostatic control of periodontal bone tissue. Periodontol 2000 2021; 86:157-187. [PMID: 33690918 DOI: 10.1111/prd.12368] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alveolar bone is a unique osseous tissue due to the proximity of dental plaque biofilms. Periodontal health and homeostasis are mediated by a balanced host immune response to these polymicrobial biofilms. Dysbiotic shifts within dental plaque biofilms can drive a proinflammatory immune response state in the periodontal epithelial and gingival connective tissues, which leads to paracrine signaling to subjacent bone cells. Sustained chronic periodontal inflammation disrupts "coupled" osteoclast-osteoblast actions, which ultimately result in alveolar bone destruction. This chapter will provide an overview of alveolar bone physiology and will highlight why the oral microbiota is a critical regulator of alveolar bone remodeling. The ecology of dental plaque biofilms will be discussed in the context that periodontitis is a polymicrobial disruption of host homeostasis. The pathogenesis of periodontal bone loss will be explained from both a historical and current perspective, providing the opportunity to revisit the role of fibrosis in alveolar bone destruction. Periodontal immune cell interactions with bone cells will be reviewed based on our current understanding of osteoimmunological mechanisms influencing alveolar bone remodeling. Lastly, probiotic and prebiotic interventions in the oral microbiota will be evaluated as potential noninvasive therapies to support alveolar bone homeostasis and prevent periodontal bone loss.
Collapse
Affiliation(s)
- Jessica D Hathaway-Schrader
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Chad M Novince
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
30
|
Low JS, Farsakoglu Y, Amezcua Vesely MC, Sefik E, Kelly JB, Harman CCD, Jackson R, Shyer JA, Jiang X, Cauley LS, Flavell RA, Kaech SM. Tissue-resident memory T cell reactivation by diverse antigen-presenting cells imparts distinct functional responses. J Exp Med 2021; 217:151854. [PMID: 32525985 PMCID: PMC7398161 DOI: 10.1084/jem.20192291] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 02/25/2020] [Accepted: 04/21/2020] [Indexed: 12/31/2022] Open
Abstract
CD8+ tissue-resident memory T cells (TRM cells) are poised at the portals of infection and provide long-term protective immunity. Despite their critical roles, the precise mechanics governing TRM cell reactivation in situ are unknown. Using a TCR-transgenic Nur77-GFP reporter to distinguish “antigen-specific” from “bystander” reactivation, we demonstrate that lung CD8+ TRM cells are reactivated more quickly, yet less efficiently, than their counterparts in the draining LNs (TLN cells). Global profiling of reactivated memory T cells revealed tissue-defined and temporally regulated recall response programs. Unlike the reactivation of CD8+ TLN cells, which is strictly dependent on CD11c+XCR1+ APCs, numerous antigen-presenting partners, both hematopoietic and non-hematopoietic, were sufficient to reactivate lung CD8+ TRM cells, but the quality of TRM cell functional responses depended on the identity of the APCs. Together, this work uncovers fundamental differences in the activation kinetics, mechanics, and effector responses between CD8+ memory T cells in peripheral vs. lymphoid organs, revealing a novel tissue-specific paradigm for the reactivation of memory CD8+ T cells.
Collapse
Affiliation(s)
- Jun Siong Low
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| | - Yagmur Farsakoglu
- NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA
| | - Maria Carolina Amezcua Vesely
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Esen Sefik
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| | - Joseph B Kelly
- Department of Ecology and Evolutionary Biology, Stony Brook University, Stony Brook, NY
| | | | - Ruaidhri Jackson
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| | - Justin A Shyer
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| | - Xiaodong Jiang
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| | - Linda S Cauley
- Department of Immunology, University of Connecticut Health Center, Farmington, CT
| | - Richard A Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT.,Howard Hughes Medical Institute, Yale University, New Haven, CT
| | - Susan M Kaech
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT.,NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA
| |
Collapse
|
31
|
Leal JM, Huang JY, Kohli K, Stoltzfus C, Lyons-Cohen MR, Olin BE, Gale M, Gerner MY. Innate cell microenvironments in lymph nodes shape the generation of T cell responses during type I inflammation. Sci Immunol 2021; 6:eabb9435. [PMID: 33579750 PMCID: PMC8274717 DOI: 10.1126/sciimmunol.abb9435] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 07/21/2020] [Accepted: 01/14/2021] [Indexed: 12/11/2022]
Abstract
Microanatomical organization of innate immune cells within lymph nodes (LNs) is critical for the generation of adaptive responses. In particular, steady-state LN-resident dendritic cells (Res cDCs) are strategically localized to intercept lymph-draining antigens. Whether myeloid cell organization changes during inflammation and how that might affect the generation of immune responses are unknown. Here, we report that during type I, but not type II, inflammation after adjuvant immunization or viral infection, antigen-presenting Res cDCs undergo CCR7-dependent intranodal repositioning from the LN periphery into the T cell zone (TZ) to elicit T cell priming. Concurrently, inflammatory monocytes infiltrate the LNs via local blood vessels, enter the TZ, and cooperate with Res cDCs by providing polarizing cytokines to optimize T cell effector differentiation. Monocyte infiltration is nonuniform across LNs, generating distinct microenvironments with varied local innate cell composition. These spatial microdomains are associated with divergent early T cell effector programming, indicating that innate microenvironments within LNs play a critical role in regulating the quality and heterogeneity of T cell responses. Together, our findings reveal that dynamic modulation of innate cell microenvironments during type I inflammation leads to optimized generation of adaptive immune responses to vaccines and infections.
Collapse
Affiliation(s)
- Joseph M Leal
- Department of Immunology, Center for Innate Immunity and Immune Disease, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Jessica Y Huang
- Department of Immunology, Center for Innate Immunity and Immune Disease, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Karan Kohli
- Department of Surgery, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Caleb Stoltzfus
- Department of Immunology, Center for Innate Immunity and Immune Disease, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Miranda R Lyons-Cohen
- Department of Immunology, Center for Innate Immunity and Immune Disease, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Brandy E Olin
- Department of Immunology, Center for Innate Immunity and Immune Disease, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Michael Gale
- Department of Immunology, Center for Innate Immunity and Immune Disease, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Michael Y Gerner
- Department of Immunology, Center for Innate Immunity and Immune Disease, University of Washington School of Medicine, Seattle, WA 98109, USA.
| |
Collapse
|
32
|
Liu L, Lin Q, Peng J, Fang J, Tan Z, Tang H, Kwan K, Nishiura K, Liang J, Kwok H, Du Z, Sun J, Liu K, Yuen KY, Wang H, Chen Z. Persistent lentivirus infection induces early myeloid suppressor cells expansion to subvert protective memory CD8 T cell response ✰,✰✰. EBioMedicine 2020; 60:103008. [PMID: 32979832 PMCID: PMC7519271 DOI: 10.1016/j.ebiom.2020.103008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 08/29/2020] [Accepted: 08/31/2020] [Indexed: 12/13/2022] Open
Abstract
Background Memory CD8+T cell responses play an essential role in protection against persistent infection. However, HIV-1 evades vaccine-induced memory CD8+T cell response by mechanisms that are not fully understood. Methods We analyzed the temporal dynamics of CD8+T cell recall activity and function during EcoHIV infection in a potent PD1-based vaccine immunized immunocompetent mice. Findings Upon intraperitoneal EcoHIV infection, high levels of HIV-1 GAG-specific CD8+T lymphocytes recall response reduced EcoHIV-infected cells significantly. However, this protective effect diminished quickly after seven days, followed by a rapid reduction of GAG-specific CD8+T cell number and activity, and viral persistence. Mechanistically, EcoHIV activated dendritic cells (DCs) and myeloid cells. Myeloid cells were infected and rapidly expanded, exhibiting elevated PD-L1/-L2 expression and T cell suppressive function before day 7, and were resistant to CD8+T cell-mediated apoptosis. Depletion of myeloid-derived suppressor cells (MDSCs) reduced EcoHIV infection and boosted T cell responses. Interpretation This study provides an overview of the temporal interplay of persistent virus, DCs, MDSCs and antigen-specific CD8+T cells during acute infection. We identify MDSCs as critical gatekeepers that restrain antiviral T cell memory responses, and highlight MDSCs as an important target for developing effective vaccines against chronic human infections. Funding Hong Kong Research Grant Council (T11–709/18-N, HKU5/CRF/13G), General Research Fund (17122915 and 17114114), Hong Kong Health and Medical Research Fund (11100752, 14130582, 16150662), Grant RGC-ANR A-HKU709/14, the San-Ming Project of Medicine (SZSM201512029), University Development Fund of the University of Hong Kong and Li Ka Shing Faculty of Medicine Matching Fund to HKU AIDS Institute.
Collapse
Affiliation(s)
- Li Liu
- AIDS Institute and Department of Microbiology, State Key Laboratory of Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong SAR, P.R. China; HKU-AIDS Institute Shenzhen Research Laboratory and AIDS Clinical Research Laboratory, Shenzhen Key Laboratory of Infection and Immunity, Shenzhen Key Clinical Department of Emerging Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, P.R. China.
| | - Qingqing Lin
- AIDS Institute and Department of Microbiology, State Key Laboratory of Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong SAR, P.R. China
| | - Jie Peng
- AIDS Institute and Department of Microbiology, State Key Laboratory of Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong SAR, P.R. China
| | - Jun Fang
- AIDS Institute and Department of Microbiology, State Key Laboratory of Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong SAR, P.R. China
| | - Zhiwu Tan
- AIDS Institute and Department of Microbiology, State Key Laboratory of Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong SAR, P.R. China
| | - Hangying Tang
- AIDS Institute and Department of Microbiology, State Key Laboratory of Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong SAR, P.R. China
| | - Kayi Kwan
- AIDS Institute and Department of Microbiology, State Key Laboratory of Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong SAR, P.R. China
| | - Kenji Nishiura
- AIDS Institute and Department of Microbiology, State Key Laboratory of Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong SAR, P.R. China
| | - Jianguo Liang
- AIDS Institute and Department of Microbiology, State Key Laboratory of Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong SAR, P.R. China
| | - Hauyee Kwok
- AIDS Institute and Department of Microbiology, State Key Laboratory of Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong SAR, P.R. China
| | - Zhenglong Du
- AIDS Institute and Department of Microbiology, State Key Laboratory of Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong SAR, P.R. China
| | - Jiaze Sun
- AIDS Institute and Department of Microbiology, State Key Laboratory of Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong SAR, P.R. China
| | - Kang Liu
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY, United States
| | - Kwok-Yung Yuen
- AIDS Institute and Department of Microbiology, State Key Laboratory of Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong SAR, P.R. China
| | - Hui Wang
- HKU-AIDS Institute Shenzhen Research Laboratory and AIDS Clinical Research Laboratory, Shenzhen Key Laboratory of Infection and Immunity, Shenzhen Key Clinical Department of Emerging Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, P.R. China.
| | - Zhiwei Chen
- AIDS Institute and Department of Microbiology, State Key Laboratory of Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong SAR, P.R. China; HKU-AIDS Institute Shenzhen Research Laboratory and AIDS Clinical Research Laboratory, Shenzhen Key Laboratory of Infection and Immunity, Shenzhen Key Clinical Department of Emerging Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, P.R. China.
| |
Collapse
|
33
|
Oliveira MMS, Westerberg LS. Cytoskeletal regulation of dendritic cells: An intricate balance between migration and presentation for tumor therapy. J Leukoc Biol 2020; 108:1051-1065. [PMID: 32557835 DOI: 10.1002/jlb.1mr0520-014rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 05/04/2020] [Accepted: 05/05/2020] [Indexed: 12/28/2022] Open
Abstract
Dendritic cells (DCs) are the main players in many approaches for cancer therapy. The idea with DC tumor therapy is to promote activation of tumor infiltrating cytotoxic T cells that kill tumor cells. This requires that DCs take up tumor Ag and present peptides on MHC class I molecules in a process called cross-presentation. For this process to be efficient, DCs have to migrate to the tumor draining lymph node and there activate the machinery for cross-presentation. In this review, we will discuss recent progress in understanding the role of actin regulators for control of DC migration and Ag presentation. The potential to target actin regulators for better DC-based tumor therapy will also be discussed.
Collapse
Affiliation(s)
- Mariana M S Oliveira
- Department of Microbiology Tumor and Cell Biology, Biomedicum, Karolinska Institutet, Stockholm, Sweden
| | - Lisa S Westerberg
- Department of Microbiology Tumor and Cell Biology, Biomedicum, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
34
|
Kidney dendritic cells: fundamental biology and functional roles in health and disease. Nat Rev Nephrol 2020; 16:391-407. [PMID: 32372062 DOI: 10.1038/s41581-020-0272-y] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2020] [Indexed: 02/06/2023]
Abstract
Dendritic cells (DCs) are chief inducers of adaptive immunity and regulate local inflammatory responses across the body. Together with macrophages, the other main type of mononuclear phagocyte, DCs constitute the most abundant component of the intrarenal immune system. This network of functionally specialized immune cells constantly surveys its microenvironment for signs of injury or infection, which trigger the initiation of an immune response. In the healthy kidney, DCs coordinate effective immune responses, for example, by recruiting neutrophils for bacterial clearance in pyelonephritis. The pro-inflammatory actions of DCs can, however, also contribute to tissue damage in various types of acute kidney injury and chronic glomerulonephritis, as DCs recruit and activate effector T cells, which release toxic mediators and maintain tubulointerstitial immune infiltrates. These actions are counterbalanced by DC subsets that promote the activation and maintenance of regulatory T cells to support resolution of the immune response and allow kidney repair. Several studies have investigated the multiple roles for DCs in kidney homeostasis and disease, but it has become clear that current tools and subset markers are not sufficient to accurately distinguish DCs from macrophages. Multidimensional transcriptomic analysis studies promise to improve mononuclear phagocyte classification and provide a clearer view of DC ontogeny and subsets.
Collapse
|
35
|
Yan LM, Lau SPN, Poh CM, Chan VSF, Chan MCW, Peiris M, Poon LLM. Heterosubtypic Protection Induced by a Live Attenuated Influenza Virus Vaccine Expressing Galactose-α-1,3-Galactose Epitopes in Infected Cells. mBio 2020; 11:e00027-20. [PMID: 32127444 PMCID: PMC7064743 DOI: 10.1128/mbio.00027-20] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 01/14/2020] [Indexed: 12/22/2022] Open
Abstract
Anti-galactose-α-1,3-galactose (anti-α-Gal) antibody is naturally expressed at a high level in humans. It constitutes about 1% of immunoglobulins found in human blood. Here, we designed a live attenuated influenza virus vaccine that can generate α-Gal epitopes in infected cells in order to facilitate opsonization of infected cells, thereby enhancing vaccine-induced immune responses. In the presence of normal human sera, cells infected with this mutant can enhance phagocytosis of human macrophages and cytotoxicity of NK cells in vitro Using a knockout mouse strain that allows expression of anti-α-Gal antibody in vivo, we showed that this strategy can increase vaccine immunogenicity and the breadth of protection. This vaccine can induce 100% protection against a lethal heterosubtypic group 1 (H5) or group 2 (mouse-adapted H3) influenza virus challenge in the mouse model. In contrast, its heterosubtypic protective effect in wild-type or knockout mice that do not have anti-α-Gal antibody expression is only partial, demonstrating that the enhanced vaccine-induced protection requires anti-α-Gal antibody upon vaccination. Anti-α-Gal-expressing knockout mice immunized with this vaccine produce robust humoral and cell-mediated responses upon a lethal virus challenge. This vaccine can stimulate CD11blo/- pulmonary dendritic cells, which are known to be crucial for clearance of influenza virus. Our approach provides a novel strategy for developing next-generation influenza virus vaccines.IMPORTANCE Influenza A viruses have multiple HA subtypes that are antigenically diverse. Classical influenza virus vaccines are subtype specific, and they cannot induce satisfactory heterosubtypic immunity against multiple influenza virus subtypes. Here, we developed a live attenuated H1N1 influenza virus vaccine that allows the expression of α-Gal epitopes by infected cells. Anti-α-Gal antibody is naturally produced by humans. In the presence of this antibody, human cells infected with this experimental vaccine virus can enhance several antibody-mediated immune responses in vitro Importantly, mice expressing anti-α-Gal antibody in vivo can be fully protected by this H1N1 vaccine against a lethal H5 or H3 virus challenge. Our work demonstrates a new strategy for using a single influenza virus strain to induce broadly cross-reactive immune responses against different influenza virus subtypes.
Collapse
Affiliation(s)
- Li-Meng Yan
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Sylvia P N Lau
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Chek Meng Poh
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Vera S F Chan
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Michael C W Chan
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Malik Peiris
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Leo L M Poon
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| |
Collapse
|
36
|
Wang L, Wang Z, Qin Y, Liang W. Delivered antigen peptides to resident CD8α+ DCs in lymph node by micelle-based vaccine augment antigen-specific CD8+ effector T cell response. Eur J Pharm Biopharm 2020; 147:76-86. [DOI: 10.1016/j.ejpb.2019.12.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Revised: 09/19/2019] [Accepted: 12/22/2019] [Indexed: 12/13/2022]
|
37
|
Olukitibi TA, Ao Z, Mahmoudi M, Kobinger GA, Yao X. Dendritic Cells/Macrophages-Targeting Feature of Ebola Glycoprotein and its Potential as Immunological Facilitator for Antiviral Vaccine Approach. Microorganisms 2019; 7:E402. [PMID: 31569539 PMCID: PMC6843631 DOI: 10.3390/microorganisms7100402] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 09/19/2019] [Accepted: 09/26/2019] [Indexed: 01/06/2023] Open
Abstract
In the prevention of epidemic and pandemic viral infection, the use of the antiviral vaccine has been the most successful biotechnological and biomedical approach. In recent times, vaccine development studies have focused on recruiting and targeting immunogens to dendritic cells (DCs) and macrophages to induce innate and adaptive immune responses. Interestingly, Ebola virus (EBOV) glycoprotein (GP) has a strong binding affinity with DCs and macrophages. Shreds of evidence have also shown that the interaction between EBOV GP with DCs and macrophages leads to massive recruitment of DCs and macrophages capable of regulating innate and adaptive immune responses. Therefore, studies for the development of vaccine can utilize the affinity between EBOV GP and DCs/macrophages as a novel immunological approach to induce both innate and acquired immune responses. In this review, we will discuss the unique features of EBOV GP to target the DC, and its potential to elicit strong immune responses while targeting DCs/macrophages. This review hopes to suggest and stimulate thoughts of developing a stronger and effective DC-targeting vaccine for diverse virus infection using EBOV GP.
Collapse
Affiliation(s)
- Titus Abiola Olukitibi
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.
| | - Zhujun Ao
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.
| | - Mona Mahmoudi
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.
| | - Gary A Kobinger
- Centre de Recherche en Infectiologie de l' Université Laval/Centre Hospitalier de l' Université Laval (CHUL), Québec, QC G1V 4G2, Canada.
| | - Xiaojian Yao
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.
| |
Collapse
|
38
|
The Role of Innate Leukocytes during Influenza Virus Infection. J Immunol Res 2019; 2019:8028725. [PMID: 31612153 PMCID: PMC6757286 DOI: 10.1155/2019/8028725] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 07/15/2019] [Indexed: 02/07/2023] Open
Abstract
Influenza virus infection is a serious threat to humans and animals, with the potential to cause severe pneumonia and death. Annual vaccination strategies are a mainstay to prevent complications related to influenza. However, protection from the emerging subtypes of influenza A viruses (IAV) even in vaccinated individuals is challenging. Innate immune cells are the first cells to respond to IAV infection in the respiratory tract. Virus replication-induced production of cytokines from airway epithelium recruits innate immune cells to the site of infection. These leukocytes, namely, neutrophils, monocytes, macrophages, dendritic cells, eosinophils, natural killer cells, innate lymphoid cells, and γδ T cells, become activated in response to IAV, to contain the virus and protect the airway epithelium while triggering the adaptive arm of the immune system. This review addresses different anti-influenza virus schemes of innate immune cells and how these cells fine-tune the balance between immunoprotection and immunopathology during IAV infection. Detailed understanding on how these innate responders execute anti-influenza activity will help to identify novel therapeutic targets to halt IAV replication and associated immunopathology.
Collapse
|
39
|
Darkwah S, Nago N, Appiah MG, Myint PK, Kawamoto E, Shimaoka M, Park EJ. Differential Roles of Dendritic Cells in Expanding CD4 T Cells in Sepsis. Biomedicines 2019; 7:biomedicines7030052. [PMID: 31323786 PMCID: PMC6783955 DOI: 10.3390/biomedicines7030052] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/12/2019] [Accepted: 07/15/2019] [Indexed: 01/01/2023] Open
Abstract
Sepsis is a systemically dysregulated inflammatory syndrome, in which dendritic cells (DCs) play a critical role in coordinating aberrant immunity. The aim of this study is to shed light on the differential roles played by systemic versus mucosal DCs in regulating immune responses in sepsis. We identified a differential impact of the systemic and mucosal DCs on proliferating allogenic CD4 T cells in a mouse model of sepsis. Despite the fact that the frequency of CD4 T cells was reduced in septic mice, septic mesenteric lymph node (MLN) DCs proved superior to septic spleen (SP) DCs in expanding allogeneic CD4 T cells. Moreover, septic MLN DCs markedly augmented the surface expression of MHC class II and CD40, as well as the messaging of interleukin-1β (IL-1β). Interestingly, IL-1β-treated CD4 T cells expanded in a dose-dependent manner, suggesting that this cytokine acts as a key mediator of MLN DCs in promoting septic inflammation. Thus, mucosal and systemic DCs were found to be functionally different in the way CD4 T cells respond during sepsis. Our study provides a molecular basis for DC activity, which can be differential in nature depending on location, whereby it induces septic inflammation or immune-paralysis.
Collapse
Affiliation(s)
- Samuel Darkwah
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Nodoka Nago
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
- Department of Clinical Nutrition, Suzuka University of Medical Science, Suzuka, Mie 510-0293, Japan
| | - Michael G Appiah
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Phyoe Kyawe Myint
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Eiji Kawamoto
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
- Department of Emergency and Disaster Medicine, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Motomu Shimaoka
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Eun Jeong Park
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan.
| |
Collapse
|
40
|
Chrisikos TT, Zhou Y, Slone N, Babcock R, Watowich SS, Li HS. Molecular regulation of dendritic cell development and function in homeostasis, inflammation, and cancer. Mol Immunol 2019; 110:24-39. [PMID: 29549977 PMCID: PMC6139080 DOI: 10.1016/j.molimm.2018.01.014] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 01/04/2018] [Accepted: 01/25/2018] [Indexed: 02/06/2023]
Abstract
Dendritic cells (DCs) are the principal antigen-presenting cells of the immune system and play key roles in controlling immune tolerance and activation. As such, DCs are chief mediators of tumor immunity. DCs can regulate tolerogenic immune responses that facilitate unchecked tumor growth. Importantly, however, DCs also mediate immune-stimulatory activity that restrains tumor progression. For instance, emerging evidence indicates the cDC1 subset has important functions in delivering tumor antigens to lymph nodes and inducing antigen-specific lymphocyte responses to tumors. Moreover, DCs control specific therapeutic responses in cancer including those resulting from immune checkpoint blockade. DC generation and function is influenced profoundly by cytokines, as well as their intracellular signaling proteins including STAT transcription factors. Regardless, our understanding of DC regulation in the cytokine-rich tumor microenvironment is still developing and must be better defined to advance cancer treatment. Here, we review literature focused on the molecular control of DCs, with a particular emphasis on cytokine- and STAT-mediated DC regulation. In addition, we highlight recent studies that delineate the importance of DCs in anti-tumor immunity and immune therapy, with the overall goal of improving knowledge of tumor-associated factors and intrinsic DC signaling cascades that influence DC function in cancer.
Collapse
Affiliation(s)
- Taylor T Chrisikos
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA; The University of Texas Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Yifan Zhou
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Natalie Slone
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA; Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Rachel Babcock
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA; The University of Texas Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Stephanie S Watowich
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA; The University of Texas Graduate School of Biomedical Sciences, Houston, TX, 77030, USA.
| | - Haiyan S Li
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
41
|
CD205-positive, Sepharose-induced peritoneal exudate cells: a new resource for DC research in the chicken. Vet Res Commun 2019; 43:115-122. [PMID: 30989431 DOI: 10.1007/s11259-019-09751-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 03/22/2019] [Indexed: 10/27/2022]
Abstract
Dendritic cells (DC) are important antigen-presenting cells and are among the least characterized immune cells in the chicken. In order to obtain chicken DC, current protocols require isolation of bone marrow myeloid progenitor cells and induction of DC differentiation with supplemental cytokines or negative selection of splenic cell preparations. Chicken peritoneal exudate cells (PEC) have traditionally been a source of various immune cells for ex vivo studies, primarily to investigate heterophils and macrophages. In this study, we observe the presence of CD205+ PEC populations, a marker of DC, as an additional resource to isolate and study chicken primary DCs. A panel of monoclonal antibodies was developed against the chicken CD205 DC marker and used to isolate CD205+ DC from the PEC population using magnetic bead cell sorting. This study reports the development of new anti-CD205 monoclonal antibodies as a reagent for chicken DC research, as well as PEC as a potential source of CD205+ DC for ex vivo studies in the chicken.
Collapse
|
42
|
Wang Z, Wang W, Chai Q, Zhu M. Langerhans Cells Control Lymphatic Vessel Function during Inflammation via LIGHT-LTβR Signaling. THE JOURNAL OF IMMUNOLOGY 2019; 202:2999-3007. [PMID: 30952816 DOI: 10.4049/jimmunol.1801578] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 03/12/2019] [Indexed: 01/03/2023]
Abstract
The lymphatic vasculature is an important route for dendritic cell (DC) or tumor cell migration from peripheral tissues to draining lymph nodes (DLNs). However, the underlying molecular and cellular mechanisms remain poorly understood. In this study, using conventional bone marrow chimeric mice and additional UVB radiation, we found that deficiency of LIGHT but not lymphotoxin (LT) α1β2, likely on radioresistant Langerhans cells (LCs), resulted in impaired skin DC migration to DLNs during LPS-induced inflammation. In addition, LT β receptor (LTβR), but not herpes virus entry mediator, was found to be the receptor of LIGHT controlling DC migration. Furthermore, conditional deficiency of LTβR in Tie2 cre or Lyve1 cre mice, but not in LTβR-deficient bone marrow chimeric mice, impaired DC migration, suggesting an important role of LTβR in radioresistant lymphatic endothelial cells (LECs), although the role of LTβR in blood endothelial cells remains intriguing. Mechanistically, the gene expression of both CCL21 and CCL19 was found to be reduced in skin LECs isolated from LC-LIGHT-conditionally deficient or Lyve1 cre Ltbr fl/fl mice compared with their controls upon LPS stimulation. Soluble recombinant LIGHT was able to upregulate CCL21 and CCL19 gene expression on SVEC4-10 endothelial cells. Doxycycline, an inhibitor of soluble LIGHT release in the inflamed skin, impaired skin CCL21 and CCL19 expression and DC migration. In addition, melanoma cell metastasis to DLNs was also inhibited in LC-LIGHT-conditionally deficient or Lyve1 cre Ltbr fl/fl mice. Together, our data suggest, to our knowledge, a previously unrecognized scenario in which LCs activate LECs via the LIGHT-LTβR signaling axis to promote DC migration or tumor cell metastasis.
Collapse
Affiliation(s)
- Zhongnan Wang
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; and.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenjun Wang
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; and.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qian Chai
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; and
| | - Mingzhao Zhu
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; and .,College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
43
|
Intravital imaging of skin infections. Cell Immunol 2019; 350:103913. [PMID: 30992120 DOI: 10.1016/j.cellimm.2019.04.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 02/11/2019] [Accepted: 04/01/2019] [Indexed: 11/23/2022]
Abstract
Intravital imaging of cutaneous immune responses has revealed intricate links between the skin's structural properties, the immune cells that reside therein, and the carefully orchestrated migratory dynamics that enable rapid sensing and subsequent elimination of skin pathogens. In particular, the development of 2-photon intravital microscopy (2P-IVM), which enables the excitation of fluorescent molecules within deep tissue with minimal light scattering and tissue damage, has proven an invaluable tool in the characterization of different cell subset's roles in skin infection. The ability to visualize cells, tissue structures, pathogens and track migratory dynamics at designated times following infection, or during inflammatory responses has been crucial in defining how immune responses in the skin are coordinated, either locally or in concert with circulating immune cells. Skin pathogens affect millions of people worldwide, and skin infections leading to cutaneous pathology have a considerable impact on the quality of life and longevity of people affected. In contrast, pathogens that infect the skin to later cause systemic illness, such as malaria parasites and a variety of arthropod-borne viruses, or infection in distant anatomical sites are a significant cause of morbidity and mortality worldwide. Here, we review recent advances and seminal studies that employed intravital imaging to characterize key immune response mechanisms in the context of viral, bacterial and parasitic skin infections, and provide insights on skin pathogens of global significance that would benefit from such investigative approaches.
Collapse
|
44
|
Park SC, Kim MJ, Baek SK, Park JH, Choi SO. Spray-Formed Layered Polymer Microneedles for Controlled Biphasic Drug Delivery. Polymers (Basel) 2019; 11:polym11020369. [PMID: 30960353 PMCID: PMC6419157 DOI: 10.3390/polym11020369] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 02/11/2019] [Accepted: 02/16/2019] [Indexed: 12/12/2022] Open
Abstract
In this study we present polymeric microneedles composed of multiple layers to control drug release kinetics. Layered microneedles were fabricated by spraying poly(lactic-co-glycolic acid) (PLGA) and polyvinylpyrrolidone (PVP) in sequence, and were characterized by mechanical testing and ex vivo skin insertion tests. The compression test demonstrated that no noticeable layer separation occurred, indicating good adhesion between PLGA and PVP layers. Histological examination confirmed that the microneedles were successfully inserted into the skin and indicated biphasic release of dyes incorporated within microneedle matrices. Structural changes of a model protein drug, bovine serum albumin (BSA), in PLGA and PVP matrices were examined by circular dichroism (CD) and fluorescence spectroscopy. The results showed that the tertiary structure of BSA was well maintained in both PLGA and PVP layers while the secondary structures were slightly changed during microneedle fabrication. In vitro release studies showed that over 60% of BSA in the PLGA layer was released within 1 h, followed by continuous slow release over the course of the experiments (7 days), while BSA in the PVP layer was completely released within 0.5 h. The initial burst of BSA from PLGA was further controlled by depositing a blank PLGA layer prior to forming the PLGA layer containing BSA. The blank PLGA layer acted as a diffusion barrier, resulting in a reduced initial burst. The formation of the PLGA diffusion barrier was visualized using confocal microscopy. Our results suggest that the spray-formed multilayer microneedles could be an attractive transdermal drug delivery system that is capable of modulating a drug release profile.
Collapse
Affiliation(s)
- Seok Chan Park
- Nanotechnology Innovation Center of Kansas State, Kansas State University, Manhattan, KS 66506, USA.
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA.
| | - Min Jung Kim
- Nanotechnology Innovation Center of Kansas State, Kansas State University, Manhattan, KS 66506, USA.
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA.
| | - Seung-Ki Baek
- QuadMedicine R&D Centre, QuadMedicine Inc., Seongnam 13209, Korea.
| | - Jung-Hwan Park
- Department of BioNano Technology, College of BioNano Technology, Gachon University, Seongnam 13120, Korea.
- Gachon BioNano Research Institute, Gachon University, Seongnam 13120, Korea.
| | - Seong-O Choi
- Nanotechnology Innovation Center of Kansas State, Kansas State University, Manhattan, KS 66506, USA.
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA.
| |
Collapse
|
45
|
Roy S, Jaeson MI, Li Z, Mahboob S, Jackson RJ, Grubor-Bauk B, Wijesundara DK, Gowans EJ, Ranasinghe C. Viral vector and route of administration determine the ILC and DC profiles responsible for downstream vaccine-specific immune outcomes. Vaccine 2019; 37:1266-1276. [PMID: 30733092 DOI: 10.1016/j.vaccine.2019.01.045] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 01/08/2019] [Accepted: 01/23/2019] [Indexed: 12/19/2022]
Abstract
This study demonstrates that route and viral vector can significantly influence the innate lymphoid cells (ILC) and dendritic cells (DC) recruited to the vaccination site, 24 h post delivery. Intranasal (i.n.) vaccination induced ST2/IL-33R+ ILC2, whilst intramuscular (i.m.) induced IL-25R+ and TSLPR+ (Thymic stromal lymphopoietin protein receptor) ILC2 subsets. However, in muscle a novel ILC subset devoid of the known ILC2 markers (IL-25R- IL-33R- TSLPR-) were found to express IL-13, unlike in lung. Different viral vectors also influenced the ILC-derived cytokines and the DC profiles at the respective vaccination sites. Both i.n. and i.m. recombinant fowlpox virus (rFPV) priming, which has been associated with induction of high avidity T cells and effective antibody differentiation exhibited low ILC2-derived IL-13, high NKp46+ ILC1/ILC3 derived IFN-γ and low IL-17A, together with enhanced CD11b+ CD103- conventional DCs (cDC). In contrast, recombinant Modified Vaccinia Ankara (rMVA) and Influenza A vector priming, which has been linked to low avidity T cells, induced opposing ILC derived-cytokine profiles and enhanced cross-presenting DCs. These observations suggested that the former ILC/DC profiles could be a predictor of a balanced cellular and humoral immune outcome. In addition, following i.n. delivery Rhinovirus (RV) and Adenovius type 5 (Ad5) vectors that induced elevated ILC2-derived IL-13, NKp46+ ILC1/ILC3-derived-IFN-γ and no IL-17A, predominantly recruited CD11b- B220+ plasmacytoid DCs (pDC). Knowing that pDC are involved in antibody differentiation, we postulate that i.n. priming with these vectors may favour induction of effective humoral immunity. Our data also revealed that vector-specific replication status and/or presence or absence of immune evasive genes can significantly alter the ILC and DC activity. Collectively, our findings suggest that understanding the route- and vector-specific ILC and DC profiles at the vaccination site may help tailor/design more efficacious viral vector-based vaccines, according to the pathogen of interest.
Collapse
Affiliation(s)
- S Roy
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology and infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra ACT 2601, Australia
| | - M I Jaeson
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology and infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra ACT 2601, Australia
| | - Z Li
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology and infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra ACT 2601, Australia
| | - S Mahboob
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology and infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra ACT 2601, Australia
| | - R J Jackson
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology and infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra ACT 2601, Australia
| | - B Grubor-Bauk
- Virology Group, Basil Hetzel Institute for Translational Health Research, University of Adelaide, Australia
| | - D K Wijesundara
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology and infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra ACT 2601, Australia; Virology Group, Basil Hetzel Institute for Translational Health Research, University of Adelaide, Australia
| | - E J Gowans
- Virology Group, Basil Hetzel Institute for Translational Health Research, University of Adelaide, Australia
| | - C Ranasinghe
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology and infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra ACT 2601, Australia.
| |
Collapse
|
46
|
Rodrigues M, Kosaric N, Bonham CA, Gurtner GC. Wound Healing: A Cellular Perspective. Physiol Rev 2019; 99:665-706. [PMID: 30475656 PMCID: PMC6442927 DOI: 10.1152/physrev.00067.2017] [Citation(s) in RCA: 1558] [Impact Index Per Article: 259.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 06/25/2018] [Accepted: 06/28/2018] [Indexed: 02/08/2023] Open
Abstract
Wound healing is one of the most complex processes in the human body. It involves the spatial and temporal synchronization of a variety of cell types with distinct roles in the phases of hemostasis, inflammation, growth, re-epithelialization, and remodeling. With the evolution of single cell technologies, it has been possible to uncover phenotypic and functional heterogeneity within several of these cell types. There have also been discoveries of rare, stem cell subsets within the skin, which are unipotent in the uninjured state, but become multipotent following skin injury. Unraveling the roles of each of these cell types and their interactions with each other is important in understanding the mechanisms of normal wound closure. Changes in the microenvironment including alterations in mechanical forces, oxygen levels, chemokines, extracellular matrix and growth factor synthesis directly impact cellular recruitment and activation, leading to impaired states of wound healing. Single cell technologies can be used to decipher these cellular alterations in diseased states such as in chronic wounds and hypertrophic scarring so that effective therapeutic solutions for healing wounds can be developed.
Collapse
Affiliation(s)
- Melanie Rodrigues
- Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Nina Kosaric
- Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Clark A Bonham
- Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Geoffrey C Gurtner
- Department of Surgery, Stanford University School of Medicine , Stanford, California
| |
Collapse
|
47
|
|
48
|
Benhadou F, Mintoff D, Del Marmol V. Psoriasis: Keratinocytes or Immune Cells - Which Is the Trigger? Dermatology 2018; 235:91-100. [PMID: 30566935 DOI: 10.1159/000495291] [Citation(s) in RCA: 155] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 11/12/2018] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Psoriasis is a common, chronic inflammatory skin disorder, which can significantly impact quality of life. Despite major breakthroughs in our understanding of the pathogenesis of psoriasis, the chronological order of the underlying mechanisms leading to the development of psoriatic plaques remains to be completely understood. SUMMARY Although psoriasis is classically perceived as a T-cell disease, it is now well recognized that T lymphocytes do not function in exclusivity. This theory is supported by evidence from transgenic murine models that develop marked psoriasiform disease. In addition, immune cells and cytokines regulate both early and late events involved in the pathogenesis of psoriasis. Key Messages: Psoriasis is a complex disease - a dynamic interplay between immune cells, keratinocytes, and various other skin-resident cells, such as endothelial and immune cells. The contribution of each cell type is crucial in the initiation and maintenance phases of psoriatic alterations.
Collapse
Affiliation(s)
- Farida Benhadou
- Dermatology Department, Erasme Hospital, Université Libre de Bruxelles - ULB, Brussels, Belgium, .,Laboratory of Stem Cells and Cancer, Unversité Libre de Bruxelles - ULB, Brussels, Belgium,
| | - Dillon Mintoff
- Dermatology Department, Sir Paul Boffa Hospital, Floriana, Malta
| | - Véronique Del Marmol
- Dermatology Department, Erasme Hospital, Université Libre de Bruxelles - ULB, Brussels, Belgium
| |
Collapse
|
49
|
Kotov DI, Pengo T, Mitchell JS, Gastinger MJ, Jenkins MK. Chrysalis: A New Method for High-Throughput Histo-Cytometry Analysis of Images and Movies. THE JOURNAL OF IMMUNOLOGY 2018; 202:300-308. [PMID: 30510065 DOI: 10.4049/jimmunol.1801202] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 11/02/2018] [Indexed: 12/17/2022]
Abstract
Advances in imaging have led to the development of powerful multispectral, quantitative imaging techniques, like histo-cytometry. The utility of this approach is limited, however, by the need for time consuming manual image analysis. We therefore developed the software Chrysalis and a group of Imaris Xtensions to automate this process. The resulting automation allowed for high-throughput histo-cytometry analysis of three-dimensional confocal microscopy and two-photon time-lapse images of T cell-dendritic cell interactions in mouse spleens. It was also applied to epi-fluorescence images to quantify T cell localization within splenic tissue by using a "signal absorption" strategy that avoids computationally intensive distance measurements. In summary, this image processing and analysis software makes histo-cytometry more useful for immunology applications by automating image analysis.
Collapse
Affiliation(s)
- Dmitri I Kotov
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455; .,Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455
| | - Thomas Pengo
- University of Minnesota Informatics Institute, University of Minnesota Twin Cities, Minneapolis, MN 55455
| | - Jason S Mitchell
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455.,University Imaging Centers, University of Minnesota, Minneapolis, MN 55455.,Department of Medicine, University of Minnesota, Minneapolis, MN 55455; and
| | | | - Marc K Jenkins
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455.,Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455
| |
Collapse
|
50
|
Huber A, Dammeijer F, Aerts JGJV, Vroman H. Current State of Dendritic Cell-Based Immunotherapy: Opportunities for in vitro Antigen Loading of Different DC Subsets? Front Immunol 2018; 9:2804. [PMID: 30559743 PMCID: PMC6287551 DOI: 10.3389/fimmu.2018.02804] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 11/14/2018] [Indexed: 12/12/2022] Open
Abstract
Dendritic cell (DC) based cancer immunotherapy aims at the activation of the immune system, and in particular tumor-specific cytotoxic T lymphocytes (CTLs) to eradicate the tumor. DCs represent a heterogeneous cell population, including conventional DCs (cDCs), consisting of cDC1s, cDC2s, plasmacytoid DCs (pDCs), and monocyte-derived DCs (moDCs). These DC subsets differ both in ontogeny and functional properties, such as the capacity to induce CD4+ and CD8+ T-cell activation. MoDCs are most frequently used for vaccination purposes, based on technical aspects such as availability and in vitro expansion. However, whether moDCs are superior over other DC subsets in inducing anti-tumor immune responses, is unknown, and likely depends on tumor type and composition of the tumor microenvironment. In this review, we discuss cellular aspects essential for DC vaccination efficacy, and the most recent findings on different DC subsets that could be used for DC-based cancer immunotherapy. This can prove valuable for the future design of more effective DC vaccines by choosing different DC subsets, and sheds light on the working mechanism of DC immunotherapy.
Collapse
Affiliation(s)
- Anne Huber
- Department of Pulmonary Medicine, Erasmus Medical Center, Rotterdam, Netherlands
| | - Floris Dammeijer
- Department of Pulmonary Medicine, Erasmus Medical Center, Rotterdam, Netherlands
- Erasmus Cancer Institute, Erasmus Medical Center, Rotterdam, Netherlands
| | - Joachim G. J. V. Aerts
- Department of Pulmonary Medicine, Erasmus Medical Center, Rotterdam, Netherlands
- Erasmus Cancer Institute, Erasmus Medical Center, Rotterdam, Netherlands
| | - Heleen Vroman
- Department of Pulmonary Medicine, Erasmus Medical Center, Rotterdam, Netherlands
- Erasmus Cancer Institute, Erasmus Medical Center, Rotterdam, Netherlands
| |
Collapse
|