1
|
Borges TJ, Lee CAA, Mucciarone K, Lima K, Lape IT, Lima-Filho M, Ayoama B, Kollar B, Gassen RB, Bonorino C, Talbot SG, Pomahac B, Lian CG, Murphy GF, Riella LV. Human type 1 conventional dendritic cells contribute to skin transplant rejection. Am J Transplant 2025:S1600-6135(25)00221-7. [PMID: 40286910 DOI: 10.1016/j.ajt.2025.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 04/02/2025] [Accepted: 04/16/2025] [Indexed: 04/29/2025]
Abstract
The skin is the most immunogenic tissue in transplantation and the most difficult tissue in which to induce immune modulation. Batf3-dependent type 1 conventional dendritic cells (cDC1s) are important in initiating rejection in murine skin transplantation. In humans, the CD141+ cDC1 subset is the functional counterpart of the murine Batf3-dependent cDC1s. However, their contribution to the rejection of human skin allografts remains unknown. Using samples from human face and upper extremity transplant recipients, we demonstrated that CD141+ cDC1s are increased and more activated in human skin grafts than native skin tissue from the same individual. Moreover, circulating and tissue CD141+ cDC1s were elevated at rejection time points. Local modulation of graft CD141+ cDC1s decreased HLA-DR expression and increased regulatory T cells, which correlated with a decreased presence of skin allogeneic T cells in a humanized transplantation model. Thus, CD141+ cDC1s play an important role in rejecting human skin allografts, and their local modulation is a promising therapeutic approach.
Collapse
Affiliation(s)
- Thiago J Borges
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| | - Catherine A A Lee
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Kyla Mucciarone
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Karina Lima
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Isadora T Lape
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Mauricio Lima-Filho
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Bruno Ayoama
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Branislav Kollar
- Department of Plastic and Hand Surgery, University of Freiburg Medical Center, Medical Faculty of the University of Freiburg, Freiburg, Germany
| | - Rodrigo B Gassen
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Cristina Bonorino
- Immunotherapy Laboratory - (LAIT) - Department of Basic Health Sciences of Federal University of Health Sciences of Porto Alegre, UFCSPA, Porto Alegre, Brazil
| | - Simon G Talbot
- Division of Plastic and Reconstructive Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Bohdan Pomahac
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Yale New Haven Hospital, Yale School of Medicine, New Haven, Connecticut, USA
| | - Christine G Lian
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - George F Murphy
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Leonardo V Riella
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
2
|
Li Y, Lu W, Xia F, Deng Y, Jin X, Xuan Y, Wang Y, Shen L, Wan J, Zhang H, Li Y, Li X, Huang L, Zhang Z. SIRPα + CD209 + cell: a specialized antigen-presenting cell that contributes to anti-SIRPα/RT therapy in colorectal cancer. Cancer Immunol Immunother 2025; 74:167. [PMID: 40208335 PMCID: PMC11985876 DOI: 10.1007/s00262-025-04025-z] [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: 12/18/2024] [Accepted: 03/17/2025] [Indexed: 04/11/2025]
Abstract
OBJECTIVE Colorectal cancer (CRC) is a leading cause of cancer-related mortality, with a need for improved treatment strategies. Antigen-presenting cells (APCs) have emerged as important modulators of immune responses in the tumor microenvironment (TME). This study aimed to explore the role of these cells in CRC and their potential synergy with radiation therapy (RT). METHODS Single-cell sequencing was performed before and after neoadjuvant therapy (NAT) to identify changes in myeloid cells within the tumor microenvironment, which was compared with peripheral blood of the same patients. The effect of RT with/without immunotherapy on these cells was evaluated in vivo and in vitro. RESULTS Single-cell sequencing showed that SIRPα + CD209 + cells are specialized antigen-presenting cells which are found to decrease in the TME while increasing in the peripheral blood after NAT. In vitro study confirmed their resistance to RT with further upregulated SIRPα expression and enhanced antigen presentation capability induced by RT. Moreover, these cells are involved in the superior tumor control by combination of RT and anti-SIRPα treatment. CONCLUSION SIRPα + CD209 + APCs play a pivotal role in CRC immune modulation and show potential for synergy with RT. These cells could be a biomarker for antigen-presenting capacity, and enhancing their APC function could potentially improve RT/PD1 effectiveness by combination with anti-SIRPα in CRC.
Collapse
Affiliation(s)
- Yida Li
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Radiation Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Weiqing Lu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Radiation Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Fan Xia
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Radiation Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Yun Deng
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Radiation Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Xin Jin
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Radiation Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Yan Xuan
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Radiation Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Yaqi Wang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Radiation Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Lijun Shen
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Radiation Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Juefeng Wan
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Radiation Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Hui Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Radiation Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Yaqi Li
- Department of Radiation Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Xinxiang Li
- Department of Radiation Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Lili Huang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Radiation Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China.
| | - Zhen Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Radiation Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China.
| |
Collapse
|
3
|
Rodriguez LIL, Amadio R, Piperno GM, Benvenuti F. Tissue-specific properties of type 1 dendritic cells in lung cancer: implications for immunotherapy. J Immunother Cancer 2025; 13:e010547. [PMID: 40132908 PMCID: PMC11938230 DOI: 10.1136/jitc-2024-010547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 02/22/2025] [Indexed: 03/27/2025] Open
Abstract
Checkpoint inhibitors have led to remarkable benefits in non-small cell lung cancer (NSCLC), yet response rates remain below expectations. High-dimensional analysis and mechanistic experiments in clinical samples and relevant NSCLC models uncovered the immune composition of lung cancer tissues, providing invaluable insights into the functional properties of tumor-infiltrating T cells and myeloid cells. Among myeloid cells, type 1 conventional dendritic cells (cDC1s) stand out for their unique ability to induce effector CD8 T cells against neoantigens and coordinate antitumoral immunity. Notably, lung resident cDC1 are particularly abundant and long-lived and express a unique tissue-specific gene program, underscoring their central role in lung immunity. Here, we discuss recent insights on the induction and regulation of antitumoral T cell responses in lung cancer, separating it from the tissue-agnostic knowledge generated from heterogeneous tumor models. We focus on the most recent studies dissecting functional states and spatial distribution of lung cDC1 across tumor stages and their impact on T cell responses to neoantigens. Finally, we highlight relevant gaps and emerging strategies to harness lung cDC1 immunostimulatory potential.
Collapse
Affiliation(s)
| | - Roberto Amadio
- Cellular Immunology, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
- Department of Life Sciences (DSV), University of Trieste, Trieste, Italy
| | - Giulia Maria Piperno
- Cellular Immunology, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Federica Benvenuti
- Cellular Immunology, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| |
Collapse
|
4
|
Li N, Steiger S, Guo Y, Li M, Wen Z, Huang M, Xie C, Jiang S, Zhang D, Zhao Y, Yu L, Wang X, Zheng Z, Zhao ZJ, Chen Y. Tubular epithelial cell-derived Flt3L is required for type 1 conventional dendritic cell (cDC1) activation and expansion in promoting the recovery in acute kidney injury. J Adv Res 2025:S2090-1232(25)00134-1. [PMID: 40023248 DOI: 10.1016/j.jare.2025.02.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 02/05/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025] Open
Abstract
INTRODUCTION Dendritic cells (DCs) play a crucial role in the recovery following acute kidney injury (AKI). Fms-related tyrosine kinase 3 ligand (Flt3L) is essential for the generation and maintenance of DCs. However, the cellular source of Flt3L in the kidney and its contribution on renal DC function during AKI remain unclear. METHODS An online available dataset and specimens collected from AKI were used to analyze FLT3L expression. Wild type (WT) mice, T cell-deficient (TcraKO), and type 1 conventional DC (cDC1)-deficient (Irf8KO) mice underwent ischemia-reperfusion (IR) injury to induce AKI. These mice were treated with either mouse recombinant Flt3L (rFlt3L) or the Flt3 inhibitor gilteritinib. In vitro, experiments with human and murine bone marrow (BM) cells, HK-2 cell line, Jurkat T cells, the monocyte cell line THP1, CD4+ T cells and cDC1s were conducted to validate the link between Flt3L and DCs. RESULTS Circulating FLT3L levels were significantly elevated in patients with AKI. This correlated with the degree of kidney dysfunction observed in these patients. Flt3L was expressed in and released by tubular epithelial cells, with minimal expression in immune cells. Flt3L primarily promoted the activation and expansion of cDC1s and polarization of CD4+T cells in vitro, an effect that was blocked by dephosphorylation of AKT and ERK signaling with gilteritinib. In vivo, gilteritinib worsened the outcomes after AKI by decreasing kidney cDC1s expansion. Conversely, therapeutic administration of rFlt3L promoted renal cDC1 accumulation and improved kidney function in mice with AKI. However, in Irf8KO mice, rFlt3 failed to improve outcomes. CONCLUSION Flt3L is upregulated in both humans and mice during IRI-induced AKI and is likely produced by tubular epithelial cells. It mainly promotes the expansion and activation of kidney cDC1 cells, thereby reducing the severity of AKI in mice. These findings suggest that Flt3L-dependent, cDC1-targeted immunotherapy could be a promising strategy for treating AKI.
Collapse
Affiliation(s)
- Na Li
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China
| | - Stefanie Steiger
- Division of Nephrology, Department of Medicine IV, University Hospital, Ludwig Maximilian University of Munich, Munich 80336, Germany
| | - Yao Guo
- Scientific Research Center, Edmond H. Fischer Translational Medical Research Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China
| | - Muzheng Li
- Department of Cardiology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China
| | - Zheqi Wen
- Department of Cardiovascular Diseases, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Mingcheng Huang
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China
| | - Chuyu Xie
- Emergency and Disaster Medical Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China
| | - Shan Jiang
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China; Scientific Research Center, Edmond H. Fischer Translational Medical Research Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China
| | - Dengyang Zhang
- Scientific Research Center, Edmond H. Fischer Translational Medical Research Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China
| | - Yuming Zhao
- Scientific Research Center, Edmond H. Fischer Translational Medical Research Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China
| | - Liuting Yu
- Scientific Research Center, Edmond H. Fischer Translational Medical Research Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China
| | - Xiaohua Wang
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China
| | - Zhihua Zheng
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China
| | - Zhizhuang Joe Zhao
- Department of Pathology, University of Oklahoma Health Sciences Center, OK 73019, USA
| | - Yun Chen
- Scientific Research Center, Edmond H. Fischer Translational Medical Research Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China.
| |
Collapse
|
5
|
Toma AI, Shah D, Roth D, Piña JO, Hymel L, Turner T, Kamalakar A, Liu K, Bartsch P, Jacobs L, D'Souza R, Liotta D, Botchwey E, Willett NJ, Goudy SL. Accelerating Oral Wound Healing Using Bilayer Biomaterial Delivery of FTY720 Immunotherapy. Adv Healthc Mater 2024; 13:e2401480. [PMID: 39388502 PMCID: PMC11616256 DOI: 10.1002/adhm.202401480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 09/27/2024] [Indexed: 10/12/2024]
Abstract
Orofacial clefts are the most common congenital craniofacial anomaly. Adverse healing following cleft palate repair can lead to oronasal fistula (ONF), a persistent connection between the oral and nasal cavities. Although human allograft tissues are currently the gold standard for ONF repair, these methods carry risks of infection and rejection, often requiring surgical revision. Immunoregenerative therapies present a novel alternative approach to harness the body's immune response and enhance the wound healing environment. An FDA-approved immunomodulatory drug, FTY720, is repurposed to reduce lymphocyte egress and induce immune cell fate switching toward pro-regenerative phenotypes. In this study, a bilayer biomaterial system is engineered using Tegaderm to secure and control the delivery of FTY720-nanofiber scaffolds (FTY720-NF). The release kinetics of the bilayer FTY720-NF is optimized to maintain drug release for up to 7 days, ensuring safe transdermal absorption and tissue biodistribution. The comprehensive immunophenotyping results demonstrate a regenerative state transition in hybrid immune cells recruited to the wound site. Further, histological evaluations reveal a significant ONF closure in mice by day 7 following bilayer FTY720-NF implantation. These findings demonstrate the utility of immunomodulatory strategies for oral wound healing, better positing the field to develop more efficacious treatment options in pediatric patients.
Collapse
Affiliation(s)
- Afra I. Toma
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyEmory UniversityAtlantaGA30322USA
- Department of Pediatrics and OtolaryngologyChildren's Healthcare of AtlantaAtlantaGA30329USA
| | - Daniel Shah
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyEmory UniversityAtlantaGA30322USA
| | - Daniela Roth
- Eunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of HealthBethesdaMD20892USA
| | - Jeremie Oliver Piña
- Eunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of HealthBethesdaMD20892USA
| | - Lauren Hymel
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyEmory UniversityAtlantaGA30322USA
| | - Thomas Turner
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyEmory UniversityAtlantaGA30322USA
| | - Archana Kamalakar
- Department of Pediatrics and OtolaryngologyChildren's Healthcare of AtlantaAtlantaGA30329USA
| | - Ken Liu
- Department of ChemistryEmory UniversityAtlantaGA30322USA
| | - Perry Bartsch
- Department of ChemistryEmory UniversityAtlantaGA30322USA
| | - Leon Jacobs
- Department of ChemistryEmory UniversityAtlantaGA30322USA
| | - Rena D'Souza
- Eunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of HealthBethesdaMD20892USA
| | - Dennis Liotta
- Department of ChemistryEmory UniversityAtlantaGA30322USA
| | - Edward Botchwey
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyEmory UniversityAtlantaGA30322USA
| | - Nick J. Willett
- Phil and Penny Knight Campus for Accelerating Scientific ImpactUniversity of OregonEugeneOR97403USA
| | - Steven L. Goudy
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of TechnologyEmory UniversityAtlantaGA30322USA
- Department of Pediatrics and OtolaryngologyChildren's Healthcare of AtlantaAtlantaGA30329USA
| |
Collapse
|
6
|
Shan Q, Qiu J, Dong Z, Xu X, Zhang S, Ma J, Liu S. Lung Immune Cell Niches and the Discovery of New Cell Subtypes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405490. [PMID: 39401416 PMCID: PMC11615829 DOI: 10.1002/advs.202405490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 08/31/2024] [Indexed: 12/06/2024]
Abstract
Immune cells in the lungs are important for maintaining lung function. The importance of immune cells in defending against lung diseases and infections is increasingly recognized. However, a primary knowledge gaps in current studies of lung immune cells is the understanding of their subtypes and functional heterogeneity. Increasing evidence supports the existence of novel immune cell subtypes that engage in the complex crosstalk between lung-resident immune cells, recruited immune cells, and epithelial cells. Therefore, further studies on how immune cells respond to perturbations in the pulmonary microenvironment are warranted. This review explores the processes behind the formation of the immune cell niche during lung development, and the characteristics and cell interaction modes of several major lung-resident immune cells. It indicates that distinct lung microenvironments or inflammatory niches can mediate the formation of different cell subtypes. These findings summarize and clarify paths to identify new cell subtypes that originate from resident progenitor cells and recruited peripheral cells, which are remodeled by the pulmonary microenvironment. The development of new techniques combining transcriptome analysis and location information is essential for identifying new immune cell subtypes and their relative immune niches, as well as for uncovering the molecular mechanisms of immune cell-mediated lung homeostasis.
Collapse
Affiliation(s)
- Qing'e Shan
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanShandong250117P. R. China
- State Key Laboratory of Environmental Chemistry and EcotoxicologyResearch Center for Eco‐Environmental SciencesChinese Academy of SciencesBeijing100085P. R. China
- School of Public HealthShandong First Medical University & Shandong Academy of Medical SciencesJinanShandong250117P. R. China
| | - Jiahuang Qiu
- Dongguan Key Laboratory of Environmental MedicineSchool of Public HealthGuangdong Medical UniversityDongguan523808P. R. China
| | - Zheng Dong
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanShandong250117P. R. China
- School of Public HealthShandong First Medical University & Shandong Academy of Medical SciencesJinanShandong250117P. R. China
| | - Xiaotong Xu
- State Key Laboratory of Environmental Chemistry and EcotoxicologyResearch Center for Eco‐Environmental SciencesChinese Academy of SciencesBeijing100085P. R. China
- School of Environmental SciencesUniversity of Chinese Academy of SciencesBeijing100049P. R. China
| | - Shuping Zhang
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanShandong250117P. R. China
- School of Public HealthShandong First Medical University & Shandong Academy of Medical SciencesJinanShandong250117P. R. China
| | - Juan Ma
- State Key Laboratory of Environmental Chemistry and EcotoxicologyResearch Center for Eco‐Environmental SciencesChinese Academy of SciencesBeijing100085P. R. China
- School of Environmental SciencesUniversity of Chinese Academy of SciencesBeijing100049P. R. China
| | - Sijin Liu
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanShandong250117P. R. China
- State Key Laboratory of Environmental Chemistry and EcotoxicologyResearch Center for Eco‐Environmental SciencesChinese Academy of SciencesBeijing100085P. R. China
- School of Public HealthShandong First Medical University & Shandong Academy of Medical SciencesJinanShandong250117P. R. China
- School of Environmental SciencesUniversity of Chinese Academy of SciencesBeijing100049P. R. China
| |
Collapse
|
7
|
Andrusaite A, Lewis J, Frede A, Farthing A, Kästele V, Montgomery J, Mowat A, Mann E, Milling S. Microbiota-derived butyrate inhibits cDC development via HDAC inhibition, diminishing their ability to prime T cells. Mucosal Immunol 2024; 17:1199-1211. [PMID: 39142634 PMCID: PMC11631772 DOI: 10.1016/j.mucimm.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/12/2024] [Accepted: 08/07/2024] [Indexed: 08/16/2024]
Abstract
Conventional dendritic cells (cDC) are central to maintaining the balance between protective immune responses and tolerance to harmless antigens, especially in the intestine. Short chain fatty acids (SCFAs) such as butyrate play critical roles in regulating intestinal immunity, but the underlying mechanisms remain unclear. Here we demonstrate that microbiota-derived butyrate alters intestinal cDC populations in vivo resulting in decreased numbers of the cDC2 lineage. By establishing a novel in vitro culture model, we show that butyrate has a direct and selective ability to repress the development of cDC2 from cDC precursors, an effect that is independent of G-protein coupled receptors (GPCRs) and is due to inhibition of histone deacetylase 3. Finally, cDC derived from pre-cDC in the presence of butyrate in vitro express lower levels of costimulatory molecules and have a decreased ability to prime naïve T cells. Together, our data show that butyrate affects the developmental trajectory of cDC, selectively repressing the cDC2 lineage and reducing their ability to stimulate T cells. These properties may help explain the ability of butyrate to maintain homeostasis in the intestine.
Collapse
Affiliation(s)
- Anna Andrusaite
- School of Infection and Immunity, University of Glasgow, UK.
| | - Jennifer Lewis
- School of Infection and Immunity, University of Glasgow, UK
| | - Annika Frede
- School of Infection and Immunity, University of Glasgow, UK
| | | | - Verena Kästele
- School of Infection and Immunity, University of Glasgow, UK
| | | | - Allan Mowat
- School of Infection and Immunity, University of Glasgow, UK
| | - Elizabeth Mann
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, UK
| | - Simon Milling
- School of Infection and Immunity, University of Glasgow, UK.
| |
Collapse
|
8
|
Lubin R, Patel AA, Mackerodt J, Zhang Y, Gvili R, Mulder K, Dutertre CA, Jalali P, Glanville JR, Hazan I, Sridharan N, Rivkin G, Akarca A, Marafioti T, Gilroy DW, Kandel L, Mildner A, Wilensky A, Asquith B, Ginhoux F, Macallan D, Yona S. The lifespan and kinetics of human dendritic cell subsets and their precursors in health and inflammation. J Exp Med 2024; 221:e20220867. [PMID: 39417994 PMCID: PMC11488382 DOI: 10.1084/jem.20220867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/16/2024] [Accepted: 09/13/2024] [Indexed: 10/19/2024] Open
Abstract
Dendritic cells (DC) are specialized mononuclear phagocytes that link innate and adaptive immunity. They comprise two principal subsets: plasmacytoid DC (pDC) and conventional DC (cDC). Understanding the generation, differentiation, and migration of cDC is critical for immune homeostasis. Through human in vivo deuterium-glucose labeling, we observed the rapid appearance of AXL+ Siglec6+ DC (ASDC) in the bloodstream. ASDC circulate for ∼2.16 days, while cDC1 and DC2 circulate for ∼1.32 and ∼2.20 days, respectively, upon release from the bone marrow. Interestingly, DC3, a cDC subset that shares several similarities with monocytes, exhibits a labeling profile closely resembling that of DC2. In a human in vivo model of cutaneous inflammation, ASDC were recruited to the inflammatory site, displaying a distinctive effector signature. Taken together, these results quantify the ephemeral circulating lifespan of human cDC and propose functions of cDC and their precursors that are rapidly recruited to sites of inflammation.
Collapse
Affiliation(s)
- Ruth Lubin
- The Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Amit A. Patel
- Division of Medicine, University College London, London, UK
| | - Jonas Mackerodt
- Department of Infectious Disease, Imperial College London, London, UK
| | - Yan Zhang
- Institute for Infection and Immunity, St. George’s, University of London, London, UK
| | - Rotem Gvili
- The Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Kevin Mulder
- Gustave Roussy Cancer Campus, Villejuif, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée—Ligue Nationale Contre le Cancer, Villejuif, France
- Université Paris-Saclay, Gif-sur-Yvette, France
| | - Charles-Antoine Dutertre
- Gustave Roussy Cancer Campus, Villejuif, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée—Ligue Nationale Contre le Cancer, Villejuif, France
| | | | | | - Idit Hazan
- The Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Nikhila Sridharan
- The Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Gurion Rivkin
- Department of Orthopaedic Surgery, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | | | | | | | - Leonid Kandel
- Department of Orthopaedic Surgery, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Alexander Mildner
- MediCity Research Laboratory, University of Turku, Turku, Finland
- InFLAMES Research Flagship, University of Turku, Turku, Finland
| | - Asaf Wilensky
- Department of Periodontology, Faculty of Dental Medicine, Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Becca Asquith
- Department of Infectious Disease, Imperial College London, London, UK
| | - Florent Ginhoux
- Gustave Roussy Cancer Campus, Villejuif, France
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong, University School of Medicine, Shanghai, China
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore
| | - Derek Macallan
- Institute for Infection and Immunity, St. George’s, University of London, London, UK
- St. George’s University Hospitals NHS Foundation Trust, London, UK
| | - Simon Yona
- The Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| |
Collapse
|
9
|
Petit M, Weber-Delacroix E, Lanthiez F, Barthélémy S, Guillou N, Firpion M, Bonduelle O, Hume DA, Combadière C, Boissonnas A. Visualizing the spatial organization of monocytes, interstitial macrophages, and tissue-specific macrophages in situ. Cell Rep 2024; 43:114847. [PMID: 39395172 DOI: 10.1016/j.celrep.2024.114847] [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: 03/25/2024] [Revised: 08/03/2024] [Accepted: 09/23/2024] [Indexed: 10/14/2024] Open
Abstract
Tissue-resident mononuclear phagocytes (MPs) are an abundant cell population whose localization in situ reflects their identity. To enable assessment of their heterogeneity, we developed the red/green/blue (RGB)-Mac mouse based upon combinations of Cx3cr1 and Csf1r reporter transgenes, providing a complete visualization of their spatial organization in situ. 3D-multi-photon imaging for spatial mapping and spectral cytometry employing the three markers in combination distinguished tissue-associated monocytes, tissue-specific macrophages, and three subsets of connective-tissue-associated MPs, including CCR2+ monocyte-derived cell, CX3CR1+, and FOLR2+ interstitial subsets, associated with distinct sub-anatomic territories. These populations were selectively reduced by blockade of CSF1, CSF2, CCR2, and CX3CR1 and efficiently reconstitute their spatial distribution after transient myelo-ablation, suggesting an autonomous regulatory environment. Our findings emphasize the organization of the MP compartment at the sub-anatomic level under steady-state conditions, thereby providing a holistic understanding of their relative heterogeneity across different tissues.
Collapse
Affiliation(s)
- Maxime Petit
- Sorbonne Université ́, Inserm U1135, CNRS ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Eléonore Weber-Delacroix
- Sorbonne Université ́, Inserm U1135, CNRS ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - François Lanthiez
- Sorbonne Université ́, Inserm U1135, CNRS ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Sandrine Barthélémy
- Sorbonne Université ́, Inserm U1135, CNRS ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Noëlline Guillou
- Sorbonne Université ́, Inserm U1135, CNRS ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Marina Firpion
- Sorbonne Université ́, Inserm U1135, CNRS ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Olivia Bonduelle
- Sorbonne Université ́, Inserm U1135, CNRS ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - David A Hume
- Mater Research Institute-University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - Christophe Combadière
- Sorbonne Université ́, Inserm U1135, CNRS ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Alexandre Boissonnas
- Sorbonne Université ́, Inserm U1135, CNRS ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France.
| |
Collapse
|
10
|
Welch BM, Parikh SA, Kay NE, Medina KL. Profound deficiencies in mature blood and bone marrow progenitor dendritic cells in Chronic Lymphocyticcytic Leukemia patients. RESEARCH SQUARE 2024:rs.3.rs-4953853. [PMID: 39399662 PMCID: PMC11469369 DOI: 10.21203/rs.3.rs-4953853/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Chronic lymphocytic leukemia (CLL) patients are immunocompromised and highly vulnerable to serious recurrent infections. Conventional dendritic cells (cDCs) and plasmacytoid DCs (pDCs) are principal sensors of infection and are essential in orchestrating innate and adaptive immune responses to resolve infection. This study identified significant deficiencies in six functionally distinct DC subsets in blood of untreated CLL (UT-CLL) patients and selective normalization of pDCs in response to acalabrutinib (a Bruton tyrosine kinase inhibitor) therapy. DCs are continuously replenished from hematopoiesis in bone marrow (BM). Four BM developmental intermediates that give rise to cDCs and pDCs were examined and significant reductions of these were identified in UT-CLL patients supporting a precursor/progeny relationship. The deficiencies in blood DCs and BM DC progenitors were significantly associated with alterations in the Flt3/FL signaling pathway critical to DC development and function. Regarding clinical parameter, cDC subset deficiencies are associated with adverse prognostic indicators of disease progression, including IGHV mutation, CD49d, CD38, and ZAP-70 status. Importantly, UT-CLL patients with shared DC subset deficiencies had shorter time-to-first treatment (TTFT), uncovering a profound alteration in innate immunity with the potential to instruct therapeutic decision-making.
Collapse
Affiliation(s)
- Baustin M. Welch
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Neil E. Kay
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
- Division of Hematology, Mayo Clinic, Rochester, MN 55905, USA
| | - Kay L. Medina
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
11
|
Dora D, Szőcs E, Soós Á, Halasy V, Somodi C, Mihucz A, Rostás M, Mógor F, Lohinai Z, Nagy N. From bench to bedside: an interdisciplinary journey through the gut-lung axis with insights into lung cancer and immunotherapy. Front Immunol 2024; 15:1434804. [PMID: 39301033 PMCID: PMC11410641 DOI: 10.3389/fimmu.2024.1434804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 08/20/2024] [Indexed: 09/22/2024] Open
Abstract
This comprehensive review undertakes a multidisciplinary exploration of the gut-lung axis, from the foundational aspects of anatomy, embryology, and histology, through the functional dynamics of pathophysiology, to implications for clinical science. The gut-lung axis, a bidirectional communication pathway, is central to understanding the interconnectedness of the gastrointestinal- and respiratory systems, both of which share embryological origins and engage in a continuous immunological crosstalk to maintain homeostasis and defend against external noxa. An essential component of this axis is the mucosa-associated lymphoid tissue system (MALT), which orchestrates immune responses across these distant sites. The review delves into the role of the gut microbiome in modulating these interactions, highlighting how microbial dysbiosis and increased gut permeability ("leaky gut") can precipitate systemic inflammation and exacerbate respiratory conditions. Moreover, we thoroughly present the implication of the axis in oncological practice, particularly in lung cancer development and response to cancer immunotherapies. Our work seeks not only to synthesize current knowledge across the spectrum of science related to the gut-lung axis but also to inspire future interdisciplinary research that bridges gaps between basic science and clinical application. Our ultimate goal was to underscore the importance of a holistic understanding of the gut-lung axis, advocating for an integrated approach to unravel its complexities in human health and disease.
Collapse
Affiliation(s)
- David Dora
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Emőke Szőcs
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Ádám Soós
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Viktória Halasy
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Csenge Somodi
- Translational Medicine Institute, Semmelweis University, Budapest, Hungary
| | - Anna Mihucz
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Melinda Rostás
- Department of Biochemistry and Molecular Biology, University of Debrecen, Debrecen, Hungary
| | - Fruzsina Mógor
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Zoltan Lohinai
- Translational Medicine Institute, Semmelweis University, Budapest, Hungary
| | - Nándor Nagy
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
12
|
Mickael C, Sanders LA, Lee MH, Kumar R, Fonseca-Balladares D, Gandjeva A, Cautivo-Reyes K, Kassa B, Kumar S, Irwin D, Swindle D, Phang T, Stearman RS, Molofsky AB, McKee AS, Stenmark KR, Graham BB, Tuder RM. Classical dendritic cells contribute to hypoxia-induced pulmonary hypertension. FASEB J 2024; 38:e70015. [PMID: 39212294 PMCID: PMC11462638 DOI: 10.1096/fj.202400338rr] [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: 02/13/2024] [Revised: 08/06/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024]
Abstract
Pulmonary hypertension (PH) is a chronic and progressive disease with significant morbidity and mortality. It is characterized by remodeled pulmonary vessels associated with perivascular and intravascular accumulation of inflammatory cells. Although there is compelling evidence that bone marrow-derived cells, such as macrophages and T cells, cluster in the vicinity of pulmonary vascular lesions in humans and contribute to PH development in different animal models, the role of dendritic cells in PH is less clear. Dendritic cells' involvement in PH is likely since they are responsible for coordinating innate and adaptive immune responses. We hypothesized that dendritic cells drive hypoxic PH. We demonstrate that a classical dendritic cell (cDC) subset (cDC2) is increased and activated in wild-type mouse lungs after hypoxia exposure. We observe significant protection after the depletion of cDCs in ZBTB46 DTR chimera mice before hypoxia exposure and after established hypoxic PH. In addition, we find that cDC depletion is associated with a reduced number of two macrophage subsets in the lung (FolR2+ MHCII+ CCR2+ and FolR2+ MHCII+ CCR2-). We found that depleting cDC2s, but not cDC1s, was protective against hypoxic PH. Finally, proof-of-concept studies in human lungs show increased perivascular cDC2s in patients with Idiopathic Pulmonary Arterial Hypertension (IPAH). Our data points to an essential role of cDCs, particularly cDC2s, in the pathophysiology of experimental PH.
Collapse
Grants
- R01 HL142701 NHLBI NIH HHS
- R01 HL161004 NHLBI NIH HHS
- R01 AI162806 NIAID NIH HHS
- R01HL142701 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01AI162806 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- American Thoracic Society (ATS)
- K01 HL161024 NHLBI NIH HHS
- K08HL168310 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- K01HL161024 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL135872 NHLBI NIH HHS
- W81XWH2210457 U.S. Department of Defense (DOD)
- Cardiovascular Medical Research and Education Fund (CMREF)
- Actelion Pharmaceuticals (Actelion Pharmaceuticals Ltd)
- R25HL146166 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R25 HL146166 NHLBI NIH HHS
- R01NS126765 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- K08 HL168310 NHLBI NIH HHS
- United Therapeutics Corporation (Uni Ther)
- R01 NS126765 NINDS NIH HHS
- 19CDA34730030 American Heart Association (AHA)
- R01HL135872 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- PO1HL152961 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL158076 NHLBI NIH HHS
- R01 H161004 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- P01 HL152961 NHLBI NIH HHS
Collapse
Affiliation(s)
- Claudia Mickael
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Aurora, Colorado, USA
- Cardiovascular Research Laboratories, University of Colorado, Aurora, Colorado, USA
| | - Linda A. Sanders
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Aurora, Colorado, USA
- Cardiovascular Research Laboratories, University of Colorado, Aurora, Colorado, USA
| | - Michael H. Lee
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
- Lung Biology Center, Zuckerberg San Francisco General Hospital, San Francisco, California, USA
| | - Rahul Kumar
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
- Lung Biology Center, Zuckerberg San Francisco General Hospital, San Francisco, California, USA
| | - Dara Fonseca-Balladares
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
- Lung Biology Center, Zuckerberg San Francisco General Hospital, San Francisco, California, USA
| | - Aneta Gandjeva
- Cardiovascular Research Laboratories, University of Colorado, Aurora, Colorado, USA
| | - Kelly Cautivo-Reyes
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, USA
- Gilead Sciences, Foster City, California, USA
| | - Biruk Kassa
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
- Lung Biology Center, Zuckerberg San Francisco General Hospital, San Francisco, California, USA
| | - Sushil Kumar
- Cardiovascular Research Laboratories, University of Colorado, Aurora, Colorado, USA
| | - David Irwin
- Cardiovascular Research Laboratories, University of Colorado, Aurora, Colorado, USA
| | - Delaney Swindle
- Cardiovascular Research Laboratories, University of Colorado, Aurora, Colorado, USA
| | - Tzu Phang
- Section of Hematology, Oncology, and Bone Marrow Transplantation-Cellular Therapeutics (BMT-CT), Department of Pediatrics, School of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Robert S. Stearman
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep, and Occupational Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Ari B. Molofsky
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, USA
| | - Amy S. McKee
- Department of Medicine, Division of Clinical Immunology, University of Colorado, Aurora, Colorado, USA
- Department of Microbiology and Immunology and ClinImmune Cell and Gene Therapy, University of Colorado, Aurora, Colorado, USA
| | - Kurt R. Stenmark
- Cardiovascular Research Laboratories, University of Colorado, Aurora, Colorado, USA
| | - Brian B. Graham
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
- Lung Biology Center, Zuckerberg San Francisco General Hospital, San Francisco, California, USA
| | - Rubin M. Tuder
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Aurora, Colorado, USA
- Cardiovascular Research Laboratories, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
13
|
Kulkarni NA, Nanjappa SG. Advances in Dendritic-Cell-Based Vaccines against Respiratory Fungal Infections. Vaccines (Basel) 2024; 12:981. [PMID: 39340013 PMCID: PMC11435842 DOI: 10.3390/vaccines12090981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/21/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
Ever since the discovery of dendritic cells by Ralph Steinman and Zanvil Cohn in 1973, it is increasingly evident that dendritic cells are integral for adaptive immune responses, and there is an undeniable focus on them for vaccines development. Fungal infections, often thought to be innocuous, are becoming significant threats due to an increased immunocompromised or immune-suppressed population and climate change. Further, the recent COVID-19 pandemic unraveled the wrath of fungal infections and devastating outcomes. Invasive fungal infections cause significant case fatality rates ranging from 20% to 90%. Regrettably, no licensed fungal vaccines exist, and there is an urgent need for preventive and therapeutic purposes. In this review, we discuss the ontogeny, subsets, tissue distribution, and functions of lung dendritic cells. In the latter part, we summarize and discuss the studies on the DC-based vaccines against pulmonary fungal infections. Finally, we highlight some emerging potential avenues that can be incorporated for DC-based vaccines against fungal infections.
Collapse
Affiliation(s)
| | - Som G. Nanjappa
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA
| |
Collapse
|
14
|
Nagai M, Okawa T, Nakata K, Takahashi D, Miyajima R, Shiratori H, Yamanaka D, Nakamura A, Oyama C, Takahashi SI, Toyama-Sorimachi N, Suzuki K, Ohashi W, Dohi T, Kawamura YI, Hase K. Sugar and arginine facilitate oral tolerance by ensuring the functionality of tolerogenic immune cell subsets in the intestine. Cell Rep 2024; 43:114490. [PMID: 38990720 DOI: 10.1016/j.celrep.2024.114490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 05/21/2024] [Accepted: 06/26/2024] [Indexed: 07/13/2024] Open
Abstract
Although oral tolerance is a critical system in regulating allergic disorders, the mechanisms by which dietary factors regulate the induction and maintenance of oral tolerance remain unclear. To address this, we explored the differentiation and function of various immune cells in the intestinal immune system under fasting and ad libitum-fed conditions before oral ovalbumin (OVA) administration. Fasting mitigated OVA-specific Treg expansion, which is essential for oral tolerance induction. This abnormality mainly resulted from functional defects in the CX3CR1+ cells responsible for the uptake of luminal OVA and reduction of tolerogenic CD103+ dendritic cells. Eventually, fasting impaired the preventive effect of oral OVA administration on asthma and allergic rhinitis development. Specific food ingredients, namely carbohydrates and arginine, were indispensable for oral tolerance induction by activating glycolysis and mTOR signaling. Overall, prior food intake and nutritional signals are critical for maintaining immune homeostasis by inducing tolerance to ingested food antigens.
Collapse
Affiliation(s)
- Motoyoshi Nagai
- Clinical Research Advancement Section, Research Institute, National Center for Global Health and Medicine, Tokyo 162-8655, Japan; Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Tokyo 105-8512, Japan.
| | - Takuma Okawa
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Tokyo 105-8512, Japan
| | - Kazuaki Nakata
- Clinical Research Advancement Section, Research Institute, National Center for Global Health and Medicine, Tokyo 162-8655, Japan
| | - Daisuke Takahashi
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Tokyo 105-8512, Japan
| | - Reina Miyajima
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Tokyo 105-8512, Japan
| | - Hiroaki Shiratori
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Tokyo 105-8512, Japan
| | - Daisuke Yamanaka
- Department of Veterinary Medical Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Atsuo Nakamura
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Tokyo 105-8512, Japan; Dairy Science and Technology Institute, Kyodo Milk Industry Co., Hinode-machi, Nishitama-gun, Tokyo, Japan
| | - Chinatsu Oyama
- Communal Laboratory, Research Institute, National Center for Global Health and Medicine, Tokyo 162-8655, Japan
| | - Shin-Ichiro Takahashi
- Departments of Animal Sciences and Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Noriko Toyama-Sorimachi
- Division of Human Immunology, International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo (IMSUT), Tokyo, Japan
| | - Koichiro Suzuki
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Tokyo 105-8512, Japan
| | - Wakana Ohashi
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Tokyo 105-8512, Japan
| | - Taeko Dohi
- Clinical Research Advancement Section, Research Institute, National Center for Global Health and Medicine, Tokyo 162-8655, Japan; Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Tokyo 105-8512, Japan
| | - Yuki I Kawamura
- Clinical Research Advancement Section, Research Institute, National Center for Global Health and Medicine, Tokyo 162-8655, Japan
| | - Koji Hase
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Tokyo 105-8512, Japan; The Institute of Fermentation Sciences (IFeS), Faculty of Food and Agricultural Sciences, Fukushima University, Kanayagawa, Fukushima 960-1296, Japan; International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo (IMSUT), Tokyo 108-8639, Japan.
| |
Collapse
|
15
|
Dotta E, Maciola AK, Baccega T, Pasqual G. Dendritic cells steering antigen and leukocyte traffic in lymph nodes. FEBS Lett 2024. [PMID: 38997244 DOI: 10.1002/1873-3468.14982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 06/01/2024] [Accepted: 06/11/2024] [Indexed: 07/14/2024]
Abstract
Dendritic cells (DCs) play a central role in initiating and shaping the adaptive immune response, thanks to their ability to uptake antigens and present them to T cells. Once in the lymph node (LN), DCs can spread the antigen to other DCs, expanding the pool of cells capable of activating specific T-cell clones. Additionally, DCs can modulate the dynamics of other immune cells, by increasing naïve T-cell dwell time, thereby facilitating the scanning for cognate antigens, and by selectively recruiting other leukocytes. Here we discuss the role of DCs in orchestrating antigen and leukocyte trafficking within the LN, together with the implications of this trafficking on T-cell activation and commitment to effector function.
Collapse
Affiliation(s)
- Enrico Dotta
- Laboratory of Synthetic Immunology, Oncology and Immunology Section, Department of Surgery Oncology and Gastroenterology, University of Padua, Italy
| | - Agnieszka Katarzyna Maciola
- Laboratory of Synthetic Immunology, Oncology and Immunology Section, Department of Surgery Oncology and Gastroenterology, University of Padua, Italy
| | - Tania Baccega
- Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Giulia Pasqual
- Laboratory of Synthetic Immunology, Oncology and Immunology Section, Department of Surgery Oncology and Gastroenterology, University of Padua, Italy
- Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| |
Collapse
|
16
|
Wilson KR, Macri C, Villadangos JA, Mintern JD. Constitutive Flt3 signaling impacts conventional dendritic cell function. Immunol Cell Biol 2024; 102:500-512. [PMID: 38693626 DOI: 10.1111/imcb.12757] [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: 03/11/2024] [Revised: 04/08/2024] [Accepted: 04/09/2024] [Indexed: 05/03/2024]
Abstract
The development of dendritic cells (DCs) depends on signaling via the FMS-like tyrosine kinase 3 (Flt3) receptor. How Flt3 signaling impacts terminally differentiated DC function is unknown. This is important given the increasing interest in exploiting Flt3 for vaccination and tumor immunotherapy. Here, we examined DCs in mice harboring constitutively activated Flt3 (Flt3-ITD). Flt3ITD/ITD mice possessed expanded splenic DC subsets including plasmacytoid DC, conventional DC (cDC)1, cDC2, double positive (DP) cDC1 (CD11c+ CD8+ CD11b- CD103+ CD86+), noncanonical (NC) cDC1 (CD11c+ CD8+ CD11b- CD103- CD86-) and single positive (SP) cDC1 (CD11c+ CD8+ CD11b- CD103- CD86+). Outcomes of constitutive Flt3 signaling differed depending on the cDC subset examined. In comparison with wild type (WT) DCs, all Flt3ITD/ITD cDCs displayed an altered surface phenotype with changes in costimulatory molecules, major histocompatibility complex class I (MHC I) and II (MHC II). Cytokine secretion patterns, antigen uptake, antigen proteolysis and antigen presenting function differed between WT and Flt3ITD/ITD subsets, particularly cDC2. In summary, Flt3 signaling impacts the function of terminally differentiated cDCs with important consequences for antigen presentation.
Collapse
Affiliation(s)
- Kayla R Wilson
- Department of Biochemistry and Pharmacology, The University of Melbourne, Bio21 Molecular Science and Biotechnology Institute, 30 Flemington Road, Parkville, VIC, Australia
| | - Christophe Macri
- Department of Biochemistry and Pharmacology, The University of Melbourne, Bio21 Molecular Science and Biotechnology Institute, 30 Flemington Road, Parkville, VIC, Australia
| | - Jose A Villadangos
- Department of Biochemistry and Pharmacology, The University of Melbourne, Bio21 Molecular Science and Biotechnology Institute, 30 Flemington Road, Parkville, VIC, Australia
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, Australia
| | - Justine D Mintern
- Department of Biochemistry and Pharmacology, The University of Melbourne, Bio21 Molecular Science and Biotechnology Institute, 30 Flemington Road, Parkville, VIC, Australia
| |
Collapse
|
17
|
Gélineau A, Marcelin G, Ouhachi M, Dussaud S, Voland L, Manuel R, Baba I, Rouault C, Yvan-Charvet L, Clément K, Tussiwand R, Huby T, Gautier EL. Fructooligosaccharides benefits on glucose homeostasis upon high-fat diet feeding require type 2 conventional dendritic cells. Nat Commun 2024; 15:5413. [PMID: 38926424 PMCID: PMC11208547 DOI: 10.1038/s41467-024-49820-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Diet composition impacts metabolic health and is now recognized to shape the immune system, especially in the intestinal tract. Nutritional imbalance and increased caloric intake are induced by high-fat diet (HFD) in which lipids are enriched at the expense of dietary fibers. Such nutritional challenge alters glucose homeostasis as well as intestinal immunity. Here, we observed that short-term HFD induced dysbiosis, glucose intolerance and decreased intestinal RORγt+ CD4 T cells, including peripherally-induced Tregs and IL17-producing (Th17) T cells. However, supplementation of HFD-fed male mice with the fermentable dietary fiber fructooligosaccharides (FOS) was sufficient to maintain RORγt+ CD4 T cell subsets and microbial species known to induce them, alongside having a beneficial impact on glucose tolerance. FOS-mediated normalization of Th17 cells and amelioration of glucose handling required the cDC2 dendritic cell subset in HFD-fed animals, while IL-17 neutralization limited FOS impact on glucose tolerance. Overall, we uncover a pivotal role of cDC2 in the control of the immune and metabolic effects of FOS in the context of HFD feeding.
Collapse
Affiliation(s)
- Adélaïde Gélineau
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Inserm, Research Unit on Cardiovascular and Metabolic Diseases, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Geneviève Marcelin
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Inserm, Nutrition and Obesities: Systemic approaches research group, NutriOmics, Paris, France
| | - Melissa Ouhachi
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Inserm, Research Unit on Cardiovascular and Metabolic Diseases, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Sébastien Dussaud
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Inserm, Research Unit on Cardiovascular and Metabolic Diseases, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Lise Voland
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Inserm, Nutrition and Obesities: Systemic approaches research group, NutriOmics, Paris, France
| | - Raoul Manuel
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Inserm, Research Unit on Cardiovascular and Metabolic Diseases, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Ines Baba
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Inserm, Research Unit on Cardiovascular and Metabolic Diseases, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Christine Rouault
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Inserm, Nutrition and Obesities: Systemic approaches research group, NutriOmics, Paris, France
| | - Laurent Yvan-Charvet
- Institut National de la Santé et de la Recherche Médicale, Inserm, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France
| | - Karine Clément
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Inserm, Nutrition and Obesities: Systemic approaches research group, NutriOmics, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital de la Pitié-Salpêtrière, service de Nutrition, Paris, France
| | - Roxane Tussiwand
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Thierry Huby
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Inserm, Research Unit on Cardiovascular and Metabolic Diseases, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Emmanuel L Gautier
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Inserm, Research Unit on Cardiovascular and Metabolic Diseases, Hôpital de la Pitié-Salpêtrière, Paris, France.
| |
Collapse
|
18
|
Oliveira TY, Merkenschlager J, Eisenreich T, Bortolatto J, Yao KH, Gatti DM, Churchill GA, Nussenzweig MC, Breton G. Quantitative trait loci mapping provides insights into the genetic regulation of dendritic cell numbers in mouse tissues. Cell Rep 2024; 43:114296. [PMID: 38823019 PMCID: PMC11726347 DOI: 10.1016/j.celrep.2024.114296] [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: 12/07/2023] [Revised: 04/02/2024] [Accepted: 05/14/2024] [Indexed: 06/03/2024] Open
Abstract
To explore the influence of genetics on homeostatic regulation of dendritic cell (DC) numbers, we present a screen of DCs and their progenitors in lymphoid and non-lymphoid tissues in Collaborative Cross (CC) and Diversity Outbred (DO) mice. We report 30 and 71 loci with logarithm of the odds (LOD) scores >8.18 and ranging from 6.67 to 8.19, respectively. The analysis reveals the highly polygenic and pleiotropic architecture of this complex trait, including many of the previously identified genetic regulators of DC development and maturation. Two SNPs in genes potentially underlying variation in DC homeostasis, a splice variant in Gramd4 (rs235532740) and a missense variant in Orai3 (rs216659754), are confirmed by gene editing using CRISPR-Cas9. Gramd4 is a central regulator of DC homeostasis that impacts the entire DC lineage, and Orai3 regulates cDC2 numbers in tissues. Overall, the data reveal a large number of candidate genes regulating DC homeostasis in vivo.
Collapse
Affiliation(s)
- Thiago Y Oliveira
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Julia Merkenschlager
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Thomas Eisenreich
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Juliana Bortolatto
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY 10065, USA
| | - Kai-Hui Yao
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | | | | | - Michel C Nussenzweig
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA; Howard Hughes Medical Institute (HHMI), The Rockefeller University, New York, NY 10065, USA.
| | - Gaëlle Breton
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
19
|
Manchester AC, Ammons DT, Lappin MR, Dow S. Single cell transcriptomic analysis of the canine duodenum in chronic inflammatory enteropathy and health. Front Immunol 2024; 15:1397590. [PMID: 38933260 PMCID: PMC11199541 DOI: 10.3389/fimmu.2024.1397590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
Chronic inflammatory enteropathy (CIE) is a common condition in dogs causing recurrent or persistent gastrointestinal clinical signs. Pathogenesis is thought to involve intestinal mucosal inflammatory infiltrates, but histopathological evaluation of intestinal biopsies from dogs with CIE fails to guide treatment, inform prognosis, or correlate with clinical remission. We employed single-cell RNA sequencing to catalog and compare the diversity of cells present in duodenal mucosal endoscopic biopsies from 3 healthy dogs and 4 dogs with CIE. Through characterization of 35,668 cells, we identified 31 transcriptomically distinct cell populations, including T cells, epithelial cells, and myeloid cells. Both healthy and CIE samples contributed to each cell population. T cells were broadly subdivided into GZMAhigh (putatively annotated as tissue resident) and IL7Rhigh (putatively annotated as non-resident) T cell categories, with evidence of a skewed proportion favoring an increase in the relative proportion of IL7Rhigh T cells in CIE dogs. Among the myeloid cells, neutrophils from CIE samples exhibited inflammatory (SOD2 and IL1A) gene expression signatures. Numerous differentially expressed genes were identified in epithelial cells, with gene set enrichment analysis suggesting enterocytes from CIE dogs may be undergoing stress responses and have altered metabolic properties. Overall, this work reveals the previously unappreciated cellular heterogeneity in canine duodenal mucosa and provides new insights into molecular mechanisms which may contribute to intestinal dysfunction in CIE. The cell type gene signatures developed through this study may also be used to better understand the subtleties of canine intestinal physiology in health and disease.
Collapse
Affiliation(s)
- Alison C. Manchester
- Colorado State University, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Fort Collins, CO, United States
| | - Dylan T. Ammons
- Colorado State University, Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Fort Collins, CO, United States
| | - Michael R. Lappin
- Colorado State University, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Fort Collins, CO, United States
| | - Steven Dow
- Colorado State University, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Fort Collins, CO, United States
- Colorado State University, Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Fort Collins, CO, United States
| |
Collapse
|
20
|
Adams NM, Das A, Yun TJ, Reizis B. Ontogeny and Function of Plasmacytoid Dendritic Cells. Annu Rev Immunol 2024; 42:347-373. [PMID: 38941603 DOI: 10.1146/annurev-immunol-090122-041105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/30/2024]
Abstract
Plasmacytoid dendritic cells (pDCs) represent a unique cell type within the innate immune system. Their defining property is the recognition of pathogen-derived nucleic acids through endosomal Toll-like receptors and the ensuing production of type I interferon and other soluble mediators, which orchestrate innate and adaptive responses. We review several aspects of pDC biology that have recently come to the fore. We discuss emerging questions regarding the lineage affiliation and origin of pDCs and argue that these cells constitute an integral part of the dendritic cell lineage. We emphasize the specific function of pDCs as innate sentinels of virus infection, particularly their recognition of and distinct response to virus-infected cells. This essential evolutionary role of pDCs has been particularly important for the control of coronaviruses, as demonstrated by the recent COVID-19 pandemic. Finally, we highlight the key contribution of pDCs to systemic lupus erythematosus, in which therapeutic targeting of pDCs is currently underway.
Collapse
Affiliation(s)
- Nicholas M Adams
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA;
| | - Annesa Das
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA;
| | - Tae Jin Yun
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA;
| | - Boris Reizis
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA;
| |
Collapse
|
21
|
Momenilandi M, Lévy R, Sobrino S, Li J, Lagresle-Peyrou C, Esmaeilzadeh H, Fayand A, Le Floc'h C, Guérin A, Della Mina E, Shearer D, Delmonte OM, Yatim A, Mulder K, Mancini M, Rinchai D, Denis A, Neehus AL, Balogh K, Brendle S, Rokni-Zadeh H, Changi-Ashtiani M, Seeleuthner Y, Deswarte C, Bessot B, Cremades C, Materna M, Cederholm A, Ogishi M, Philippot Q, Beganovic O, Ackermann M, Wuyts M, Khan T, Fouéré S, Herms F, Chanal J, Palterer B, Bruneau J, Molina TJ, Leclerc-Mercier S, Prétet JL, Youssefian L, Vahidnezhad H, Parvaneh N, Claeys KG, Schrijvers R, Luka M, Pérot P, Fourgeaud J, Nourrisson C, Poirier P, Jouanguy E, Boisson-Dupuis S, Bustamante J, Notarangelo LD, Christensen N, Landegren N, Abel L, Marr N, Six E, Langlais D, Waterboer T, Ginhoux F, Ma CS, Tangye SG, Meyts I, Lachmann N, Hu J, Shahrooei M, Bossuyt X, Casanova JL, Béziat V. FLT3L governs the development of partially overlapping hematopoietic lineages in humans and mice. Cell 2024; 187:2817-2837.e31. [PMID: 38701783 PMCID: PMC11149630 DOI: 10.1016/j.cell.2024.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 03/04/2024] [Accepted: 04/10/2024] [Indexed: 05/05/2024]
Abstract
FMS-related tyrosine kinase 3 ligand (FLT3L), encoded by FLT3LG, is a hematopoietic factor essential for the development of natural killer (NK) cells, B cells, and dendritic cells (DCs) in mice. We describe three humans homozygous for a loss-of-function FLT3LG variant with a history of various recurrent infections, including severe cutaneous warts. The patients' bone marrow (BM) was hypoplastic, with low levels of hematopoietic progenitors, particularly myeloid and B cell precursors. Counts of B cells, monocytes, and DCs were low in the patients' blood, whereas the other blood subsets, including NK cells, were affected only moderately, if at all. The patients had normal counts of Langerhans cells (LCs) and dermal macrophages in the skin but lacked dermal DCs. Thus, FLT3L is required for B cell and DC development in mice and humans. However, unlike its murine counterpart, human FLT3L is required for the development of monocytes but not NK cells.
Collapse
Affiliation(s)
- Mana Momenilandi
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France
| | - Romain Lévy
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France; Pediatric Hematology-Immunology and Rheumatology Unit, Necker Hospital for Sick Children, AP-HP, Paris, France
| | - Steicy Sobrino
- Laboratory of Chromatin and Gene Regulation During Development, Paris Cité University, UMR1163 INSERM, Imagine Institute, Paris, France; Laboratory of Human Lymphohematopoiesis, INSERM, Imagine Institute, Paris, France
| | - Jingwei Li
- Jake Gittlen Laboratories for Cancer Research, Pennsylvania State University College of Medicine, Hershey, PA, USA; Department of Pathology and Laboratory Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Chantal Lagresle-Peyrou
- Paris Cité University, Imagine Institute, Paris, France; Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, AP-HP, INSERM, Paris, France
| | - Hossein Esmaeilzadeh
- Allergy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Allergy and Clinical Immunology, Namazi Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Antoine Fayand
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France; Sorbonne University, AP-HP, Tenon Hospital, Department of Internal Medicine, Paris, France
| | - Corentin Le Floc'h
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France
| | - Antoine Guérin
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia; St. Vincent's Clinical School, Faculty of Medicine, University of NSW, Sydney, NSW, Australia
| | - Erika Della Mina
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia; St. Vincent's Clinical School, Faculty of Medicine, University of NSW, Sydney, NSW, Australia
| | - Debra Shearer
- Jake Gittlen Laboratories for Cancer Research, Pennsylvania State University College of Medicine, Hershey, PA, USA; Department of Pathology and Laboratory Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Ottavia M Delmonte
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ahmad Yatim
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Kevin Mulder
- Gustave Roussy Cancer Campus, Villejuif, France; Paris-Saclay University, Ile-de-France, France
| | - Mathieu Mancini
- Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC, Canada; Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - Darawan Rinchai
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Adeline Denis
- Laboratory of Human Lymphohematopoiesis, INSERM, Imagine Institute, Paris, France
| | - Anna-Lena Neehus
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France
| | - Karla Balogh
- Jake Gittlen Laboratories for Cancer Research, Pennsylvania State University College of Medicine, Hershey, PA, USA; Department of Pathology and Laboratory Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Sarah Brendle
- Jake Gittlen Laboratories for Cancer Research, Pennsylvania State University College of Medicine, Hershey, PA, USA; Department of Pathology and Laboratory Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA; Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Hassan Rokni-Zadeh
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences (ZUMS), Zanjan, Iran
| | - Majid Changi-Ashtiani
- School of Mathematics, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
| | - Yoann Seeleuthner
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France
| | - Caroline Deswarte
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France
| | - Boris Bessot
- Paris Cité University, Imagine Institute, Paris, France; Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, AP-HP, INSERM, Paris, France
| | - Cassandre Cremades
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France
| | - Marie Materna
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France
| | - Axel Cederholm
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Masato Ogishi
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Quentin Philippot
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France
| | - Omer Beganovic
- Laboratoire d'Onco-hématologie, Necker Hospital for Sick Children, AP-HP, Paris, France
| | - Mania Ackermann
- Hannover Medical School, Department of Pediatric Pulmonology, Allergology and Neonatology, Hannover, Germany; Hannover Medical School, Cluster of Excellence RESIST (EXC 2155), Hannover, Germany; Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
| | - Margareta Wuyts
- Department of Microbiology and Immunology, Clinical and Diagnostic Immunology, KU Leuven, Leuven, Belgium
| | | | - Sébastien Fouéré
- Groupe Hospitalier Saint-Louis, Lariboisière, Fernand-Widal, CeGIDD, AP-HP, Paris, France
| | - Florian Herms
- Dermatology Department, Paris-Cité University, INSERM 976, Saint Louis Hospital, Paris, France
| | - Johan Chanal
- Dermatology Department, Cochin Hospital, INSERM U1016, AP-HP, Paris, France
| | - Boaz Palterer
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Julie Bruneau
- Department of Pathology, Necker Hospital for Sick Children, AP-HP, Paris-Cité University, Paris, France
| | - Thierry J Molina
- Department of Pathology, Necker Hospital for Sick Children, AP-HP, Paris-Cité University, Paris, France
| | - Stéphanie Leclerc-Mercier
- Department of Pathology, Necker Hospital for Sick Children, AP-HP, Paris-Cité University, Paris, France
| | - Jean-Luc Prétet
- Papillomavirus National Reference Center, Besançon Hospital, Besançon, France
| | - Leila Youssefian
- Department of Pathology and Laboratory Medicine, UCLA Clinical Genomics Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Hassan Vahidnezhad
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Nima Parvaneh
- Department of Pediatrics, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Kristl G Claeys
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium; Laboratory for Muscle Diseases and Neuropathies, Department of Neurosciences, KU Leuven, and Leuven Brain Institute (LBI), Leuven, Belgium
| | - Rik Schrijvers
- Department of Microbiology, Immunology and Transplantation, Allergy and Clinical Immunology Research Group, KU Leuven, Leuven, Belgium
| | - Marine Luka
- Labtech Single-Cell@Imagine, Imagine Institute, INSERM UMR 1163, 75015 Paris, France
| | - Philippe Pérot
- Pathogen Discovery Laboratory, Institut Pasteur, Paris Cité University, Paris, France
| | - Jacques Fourgeaud
- Paris Cité University, URP 7328 FETUS, Paris, France; Microbiology Department, AP-HP, Necker Hospital for Sick Children, Paris, France
| | - Céline Nourrisson
- Clermont Auvergne University, INSERM U1071, M2iSH, USC INRAE 1382, CHU Clermont-Ferrand, 3IHP, Department of Parasitology-Mycology, Clermont-Ferrand, France; National Reference Center for Cryptosporidiosis, Microsporidia and Other Digestive Protozoa, Clermont-Ferrand, France
| | - Philippe Poirier
- Clermont Auvergne University, INSERM U1071, M2iSH, USC INRAE 1382, CHU Clermont-Ferrand, 3IHP, Department of Parasitology-Mycology, Clermont-Ferrand, France; National Reference Center for Cryptosporidiosis, Microsporidia and Other Digestive Protozoa, Clermont-Ferrand, France
| | - Emmanuelle Jouanguy
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France; St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Stéphanie Boisson-Dupuis
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France; St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Jacinta Bustamante
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France; St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA; Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children, AP-HP, Paris, France
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Neil Christensen
- Jake Gittlen Laboratories for Cancer Research, Pennsylvania State University College of Medicine, Hershey, PA, USA; Department of Pathology and Laboratory Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Nils Landegren
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden; Centre for Molecular Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Laurent Abel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France; St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Nico Marr
- Research Branch, Sidra Medicine, Doha, Qatar; College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Emmanuelle Six
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, AP-HP, INSERM, Paris, France
| | - David Langlais
- Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC, Canada; Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada; Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Tim Waterboer
- Infections and Cancer Epidemiology, Infection, Inflammation and Cancer Program, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Florent Ginhoux
- Gustave Roussy Cancer Campus, Villejuif, France; Paris-Saclay University, Ile-de-France, France
| | - Cindy S Ma
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia; St. Vincent's Clinical School, Faculty of Medicine, University of NSW, Sydney, NSW, Australia
| | - Stuart G Tangye
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia; St. Vincent's Clinical School, Faculty of Medicine, University of NSW, Sydney, NSW, Australia
| | - Isabelle Meyts
- Laboratory of Inborn Errors of Immunity, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium; Department of Pediatrics, Leuven University Hospitals, Leuven, Belgium
| | - Nico Lachmann
- Hannover Medical School, Department of Pediatric Pulmonology, Allergology and Neonatology, Hannover, Germany; Hannover Medical School, Cluster of Excellence RESIST (EXC 2155), Hannover, Germany; Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
| | - Jiafen Hu
- Jake Gittlen Laboratories for Cancer Research, Pennsylvania State University College of Medicine, Hershey, PA, USA; Department of Pathology and Laboratory Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Mohammad Shahrooei
- Department of Microbiology and Immunology, Clinical and Diagnostic Immunology, KU Leuven, Leuven, Belgium; Specialized Immunology Laboratory of Dr. Shahrooei, Tehran, Iran
| | - Xavier Bossuyt
- Department of Microbiology and Immunology, Clinical and Diagnostic Immunology, KU Leuven, Leuven, Belgium; Department of Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France; Pediatric Hematology-Immunology and Rheumatology Unit, Necker Hospital for Sick Children, AP-HP, Paris, France; St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA; Howard Hughes Medical Institute, New York, NY, USA
| | - Vivien Béziat
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France; St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA.
| |
Collapse
|
22
|
Mousa M, Liang Y, Tung LT, Wang H, Krawczyk C, Langlais D, Nijnik A. Chromatin-binding deubiquitinase MYSM1 acts in haematopoietic progenitors to control dendritic cell development and to program dendritic cell responses to microbial stimulation. Immunology 2024; 172:109-126. [PMID: 38316548 DOI: 10.1111/imm.13758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 01/08/2024] [Indexed: 02/07/2024] Open
Abstract
Dendritic cells (DCs) are the most significant antigen presenting cells of the immune system, critical for the activation of naïve T cells. The pathways controlling DC development, maturation, and effector function therefore require precise regulation to allow for an effective induction of adaptive immune response. MYSM1 is a chromatin binding deubiquitinase (DUB) and an activator of gene expression via its catalytic activity for monoubiquitinated histone H2A (H2A-K119ub), which is a highly abundant repressive epigenetic mark. MYSM1 is an important regulator of haematopoiesis in mouse and human, and a systemic constitutive loss of Mysm1 in mice results in a depletion of many haematopoietic progenitors, including DC precursors, with the downstream loss of most DC lineage cells. However, the roles of MYSM1 at the later checkpoints in DC development, maturation, activation, and effector function at present remain unknown. In the current work, using a range of novel mouse models (Mysm1flCreERT2, Mysm1flCD11c-cre, Mysm1DN), we further the understanding of MYSM1 functions in the DC lineage: assessing the requirement for MYSM1 in DC development independently of other complex developmental phenotypes, exploring its role at the later checkpoints in DC maintenance and activation in response to microbial stimulation, and testing the requirement for the DUB catalytic activity of MYSM1 in these processes. Surprisingly, we demonstrate that MYSM1 expression and catalytic activity in DCs are dispensable for the maintenance of DC numbers in vivo or for DC activation in response to microbial stimulation. In contrast, MYSM1 acts via its DUB catalytic activity specifically in haematopoietic progenitors to allow normal DC lineage development, and its loss results not only in a severe DC depletion but also in the production of functionally altered DCs, with a dysregulation of many housekeeping transcriptional programs and significantly altered responses to microbial stimulation.
Collapse
Affiliation(s)
- Marwah Mousa
- Department of Physiology, McGill University, Montreal, Quebec, Canada
- McGill University Research Centre on Complex Traits, McGill University, Montreal, Quebec, Canada
| | - Yue Liang
- Department of Physiology, McGill University, Montreal, Quebec, Canada
- McGill University Research Centre on Complex Traits, McGill University, Montreal, Quebec, Canada
| | - Lin Tze Tung
- Department of Physiology, McGill University, Montreal, Quebec, Canada
- McGill University Research Centre on Complex Traits, McGill University, Montreal, Quebec, Canada
| | - HanChen Wang
- Department of Physiology, McGill University, Montreal, Quebec, Canada
- McGill University Research Centre on Complex Traits, McGill University, Montreal, Quebec, Canada
| | - Connie Krawczyk
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, Michigan, United States
| | - David Langlais
- McGill University Research Centre on Complex Traits, McGill University, Montreal, Quebec, Canada
- Department of Human Genetics, McGill University Genome Centre, McGill University, Montreal, Quebec, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
| | - Anastasia Nijnik
- Department of Physiology, McGill University, Montreal, Quebec, Canada
- McGill University Research Centre on Complex Traits, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
23
|
Minutti CM, Piot C, Pereira da Costa M, Chakravarty P, Rogers N, Huerga Encabo H, Cardoso A, Loong J, Bessou G, Mionnet C, Langhorne J, Bonnet D, Dalod M, Tomasello E, Reis e Sousa C. Distinct ontogenetic lineages dictate cDC2 heterogeneity. Nat Immunol 2024; 25:448-461. [PMID: 38351322 PMCID: PMC10907303 DOI: 10.1038/s41590-024-01745-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 01/08/2024] [Indexed: 03/03/2024]
Abstract
Conventional dendritic cells (cDCs) include functionally and phenotypically diverse populations, such as cDC1s and cDC2s. The latter population has been variously subdivided into Notch-dependent cDC2s, KLF4-dependent cDC2s, T-bet+ cDC2As and T-bet- cDC2Bs, but it is unclear how all these subtypes are interrelated and to what degree they represent cell states or cell subsets. All cDCs are derived from bone marrow progenitors called pre-cDCs, which circulate through the blood to colonize peripheral tissues. Here, we identified distinct mouse pre-cDC2 subsets biased to give rise to cDC2As or cDC2Bs. We showed that a Siglec-H+ pre-cDC2A population in the bone marrow preferentially gave rise to Siglec-H- CD8α+ pre-cDC2As in tissues, which differentiated into T-bet+ cDC2As. In contrast, a Siglec-H- fraction of pre-cDCs in the bone marrow and periphery mostly generated T-bet- cDC2Bs, a lineage marked by the expression of LysM. Our results showed that cDC2A versus cDC2B fate specification starts in the bone marrow and suggest that cDC2 subsets are ontogenetically determined lineages, rather than cell states imposed by the peripheral tissue environment.
Collapse
Affiliation(s)
- Carlos M Minutti
- Immunobiology Laboratory, The Francis Crick Institute, London, UK.
- Immunoregulation Laboratory, Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, Portugal.
| | - Cécile Piot
- Immunobiology Laboratory, The Francis Crick Institute, London, UK
| | | | - Probir Chakravarty
- Bioinformatics and Biostatistics, The Francis Crick Institute, London, UK
| | - Neil Rogers
- Immunobiology Laboratory, The Francis Crick Institute, London, UK
| | | | - Ana Cardoso
- Immunobiology Laboratory, The Francis Crick Institute, London, UK
| | - Jane Loong
- Retroviral Immunology Laboratory, The Francis Crick Institute, London, UK
| | - Gilles Bessou
- Aix-Marseille University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, Marseille, France
| | - Cyrille Mionnet
- Aix-Marseille University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, Marseille, France
| | - Jean Langhorne
- Malaria Immunology Laboratory, The Francis Crick Institute, London, UK
| | - Dominique Bonnet
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, London, UK
| | - Marc Dalod
- Aix-Marseille University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, Marseille, France
| | - Elena Tomasello
- Aix-Marseille University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, Marseille, France
| | | |
Collapse
|
24
|
Britsch S, Langer H, Duerschmied D, Becher T. The Evolving Role of Dendritic Cells in Atherosclerosis. Int J Mol Sci 2024; 25:2450. [PMID: 38397127 PMCID: PMC10888834 DOI: 10.3390/ijms25042450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/01/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Atherosclerosis, a major contributor to cardiovascular morbidity and mortality, is characterized by chronic inflammation of the arterial wall. This inflammatory process is initiated and maintained by both innate and adaptive immunity. Dendritic cells (DCs), which are antigen-presenting cells, play a crucial role in the development of atherosclerosis and consist of various subtypes with distinct functional abilities. Following the recognition and binding of antigens, DCs become potent activators of cellular responses, bridging the innate and adaptive immune systems. The modulation of specific DC subpopulations can have either pro-atherogenic or atheroprotective effects, highlighting the dual pro-inflammatory or tolerogenic roles of DCs. In this work, we provide a comprehensive overview of the evolving roles of DCs and their subtypes in the promotion or limitation of atherosclerosis development. Additionally, we explore antigen pulsing and pharmacological approaches to modulate the function of DCs in the context of atherosclerosis.
Collapse
Affiliation(s)
- Simone Britsch
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, Centre for Acute Cardiovascular Medicine Mannheim (ZKAM), University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, 69117 Mannheim, Germany; (H.L.); (D.D.); (T.B.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, 13092 Mannheim, Germany
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Harald Langer
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, Centre for Acute Cardiovascular Medicine Mannheim (ZKAM), University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, 69117 Mannheim, Germany; (H.L.); (D.D.); (T.B.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, 13092 Mannheim, Germany
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Daniel Duerschmied
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, Centre for Acute Cardiovascular Medicine Mannheim (ZKAM), University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, 69117 Mannheim, Germany; (H.L.); (D.D.); (T.B.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, 13092 Mannheim, Germany
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Tobias Becher
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, Centre for Acute Cardiovascular Medicine Mannheim (ZKAM), University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, 69117 Mannheim, Germany; (H.L.); (D.D.); (T.B.)
| |
Collapse
|
25
|
Wang H, Tao F, Li CY, Yang GJ, Chen J. Short-term administration of Qipian®, a mixed bacterial lysate, inhibits airway inflammation in ovalbumin-induced mouse asthma by modulating cellular, humoral and neurogenic immune responses. Life Sci 2024; 336:122310. [PMID: 38013140 DOI: 10.1016/j.lfs.2023.122310] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 11/29/2023]
Abstract
AIMS Qipian® is a commercialized agent composed of extracts of three genera of commensal bacteria, and its mechanism of action on asthma is unclear. This study aimed to examine the impact of Qipian® on airway inflammation and investigate the underlying mechanisms. MATERIALS AND METHODS Qipian® or dexamethasone (DEX) was administered before OVA challenge in an ovalbumin-induced asthma mouse model, and then asthmatic symptoms were observed and scored. Samples of lung tissues, blood, and bronchoalveolar lavage fluid (BALF) were collected, and eosinophils (Eos), immunoglobins (Igs), and type 1 T helper (Th1)/Th2 cell cytokines were measured. Mucus production in the lung was assessed by periodic acid-Schiff (PAS) staining. The effects of Qipian® on dendritic and T regulatory (Treg) cells were investigated using flow cytometry. KEY FINDINGS The short-term administration of Qipian® significantly inhibited the cardinal features of allergic asthma, including an elevated asthmatic behaviour score, airway inflammation and immune response. Histological analysis of the lungs showed that Qipian® attenuated airway inflammatory cell infiltration and mucus hyperproduction. Qipian® restored Th1/Th2 imbalance by decreasing interleukin (IL)-4, IL-5, and IL-13 while increasing interferon (IFN)-γ and IL-10. Further investigation revealed that Qipian® treatment induced the upregulation of CD4+CD25+Foxp3+ Treg cells and CD103+ DCs and downregulation of tachykinins neurokinin A (NKA) and NKB in the lung. SIGNIFICANCE Our findings suggested that short-term treatment with Qipian® could alleviate inflammation in allergic asthma through restoring the Th1/Th2 balance by recruiting Treg cells to airways and inducing the proliferation of CD103+ DCs, which actually provides a new treatment option for asthma.
Collapse
Affiliation(s)
- Huiying Wang
- Department of Allergy and Clinical Immunology, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310009, China.
| | - Fan Tao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang Province 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo, Zhejiang Province 315832, China.
| | - Chang-Yun Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang Province 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo, Zhejiang Province 315832, China.
| | - Guan-Jun Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang Province 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo, Zhejiang Province 315832, China.
| | - Jiong Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang Province 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo, Zhejiang Province 315832, China.
| |
Collapse
|
26
|
Toma AI, Shah D, Roth D, Oliver Piña J, Hymel L, Turner T, Kamalakar A, Liu K, Bartsch P, Jacobs L, D'Souza R, Liotta D, Botchwey E, Willett NJ, Goudy SL. Harnessing Bilayer Biomaterial Delivery of FTY720 as an Immunotherapy to Accelerate Oral Wound Healing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.22.573096. [PMID: 38187740 PMCID: PMC10769397 DOI: 10.1101/2023.12.22.573096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Orofacial clefts are the most common craniofacial congenital anomaly. Following cleft palate repair, up to 60% of surgeries have wound healing complications leading to oronasal fistula (ONF), a persistent connection between the roof of the mouth and the nasal cavity. The current gold standard methods for ONF repair use human allograft tissues; however, these procedures have risks of graft infection and/or rejection, requiring surgical revisions. Immunoregenerative therapies present a novel alternative approach to harness the body's immune response and enhance the wound healing environment. We utilized a repurposed FDA-approved immunomodulatory drug, FTY720, to reduce the egress of lymphocytes and induce immune cell fate switching toward pro-regenerative phenotypes. Here, we engineered a bilayer biomaterial system using Tegaderm™, a liquid-impermeable wound dressing, to secure and control the delivery of FTY720- nanofiber scaffolds (FTY720-NF). We optimized release kinetics of the bilayer FTY720-NF to sustain drug release for up to 7d with safe, efficacious transdermal absorption and tissue biodistribution. Through comprehensive immunophenotyping, our results illustrate a pseudotime pro-regenerative state transition in recruited hybrid immune cells to the wound site. Additional histological assessments established a significant difference in full thickness ONF closure in mice on Day 7 following treatment with bilayer FTY720-NF, compared to controls. These findings demonstrate the utility of immunomodulatory strategies for oral wound healing, better positing the field to develop more efficacious treatment options for pediatric patients. One Sentence Summary Local delivery of bilayer FTY720-nanofiber scaffolds in an ONF mouse model promotes complete wound closure through modulation of pro-regenerative immune and stromal cells.
Collapse
|
27
|
Probst HC, Stoitzner P, Amon L, Backer RA, Brand A, Chen J, Clausen BE, Dieckmann S, Dudziak D, Heger L, Hodapp K, Hornsteiner F, Hovav AH, Jacobi L, Ji X, Kamenjarin N, Lahl K, Lahmar I, Lakus J, Lehmann CHK, Ortner D, Picard M, Roberti MP, Rossnagel L, Saba Y, Schalla C, Schlitzer A, Schraml BU, Schütze K, Seichter A, Seré K, Seretis A, Sopper S, Strandt H, Sykora MM, Theobald H, Tripp CH, Zitvogel L. Guidelines for DC preparation and flow cytometry analysis of mouse nonlymphoid tissues. Eur J Immunol 2023; 53:e2249819. [PMID: 36512638 DOI: 10.1002/eji.202249819] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 08/24/2022] [Accepted: 08/25/2022] [Indexed: 12/15/2022]
Abstract
This article is part of the Dendritic Cell Guidelines article series, which provides a collection of state-of-the-art protocols for the preparation, phenotype analysis by flow cytometry, generation, fluorescence microscopy and functional characterization of mouse and human dendritic cells (DC) from lymphoid organs and various nonlymphoid tissues. DC are sentinels of the immune system present in almost every mammalian organ. Since they represent a rare cell population, DC need to be extracted from organs with protocols that are specifically developed for each tissue. This article provides detailed protocols for the preparation of single-cell suspensions from various mouse nonlymphoid tissues, including skin, intestine, lung, kidney, mammary glands, oral mucosa and transplantable tumors. Furthermore, our guidelines include comprehensive protocols for multiplex flow cytometry analysis of DC subsets and feature top tricks for their proper discrimination from other myeloid cells. With this collection, we provide guidelines for in-depth analysis of DC subsets that will advance our understanding of their respective roles in healthy and diseased tissues. 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 coauthors, making it an essential resource for basic and clinical DC immunologists.
Collapse
Affiliation(s)
- Hans Christian Probst
- Institute of Immunology, University Medical Center Mainz, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Patrizia Stoitzner
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Lukas Amon
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Hartmannstraße 14, D-91052, Erlangen, Germany
| | - Ronald A Backer
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Anna Brand
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Jianzhou Chen
- Gustave Roussy Cancer Campus (GRCC), U1015 INSERM, University Paris Saclay, Villejuif, France
| | - Björn E Clausen
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Sophie Dieckmann
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Diana Dudziak
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Hartmannstraße 14, D-91052, Erlangen, Germany
- Medical Immunology Campus Erlangen (MICE), D-91054, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Germany
- Friedrich-Alexander University (FAU), Erlangen-Nürnberg, Germany
| | - Lukas Heger
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Hartmannstraße 14, D-91052, Erlangen, Germany
| | - Katrin Hodapp
- Institute of Immunology, University Medical Center Mainz, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Florian Hornsteiner
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Avi-Hai Hovav
- Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Lukas Jacobi
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Hartmannstraße 14, D-91052, Erlangen, Germany
| | - Xingqi Ji
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, 82152, Planegg-Martinsried, Germany
- Institute for Cardiovascular Physiology and Pathophysiology, Biomedical Center, Faculty of Medicine, LMU Munich, 82152, Planegg-Martinsried, Germany
| | - Nadine Kamenjarin
- Institute of Immunology, University Medical Center Mainz, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Katharina Lahl
- Section for Experimental and Translational Immunology, Institute for Health Technology, Technical University of Denmark (DTU), Kongens Lyngby, 2800, Denmark
- Immunology Section, Lund University, Lund, 221 84, Sweden
| | - Imran Lahmar
- Gustave Roussy Cancer Campus (GRCC), U1015 INSERM, University Paris Saclay, Villejuif, France
| | - Jelena Lakus
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Christian H K Lehmann
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Hartmannstraße 14, D-91052, Erlangen, Germany
- Medical Immunology Campus Erlangen (MICE), D-91054, Erlangen, Germany
| | - Daniela Ortner
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Marion Picard
- Gustave Roussy Cancer Campus (GRCC), U1015 INSERM, University Paris Saclay, Villejuif, France
| | - Maria Paula Roberti
- Gustave Roussy Cancer Campus (GRCC), U1015 INSERM, University Paris Saclay, Villejuif, France
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD), Heidelberg, Germany
- Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lukas Rossnagel
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Hartmannstraße 14, D-91052, Erlangen, Germany
| | - Yasmin Saba
- Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Carmen Schalla
- Institute for Biomedical Engineering, Department of Cell Biology, RWTH Aachen University Medical School, Aachen, Germany
- Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Andreas Schlitzer
- Quantitative Systems Biology, Life and Medical Sciences (LIMES) Institute, University of Bonn, Germany
| | - Barbara U Schraml
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, 82152, Planegg-Martinsried, Germany
- Institute for Cardiovascular Physiology and Pathophysiology, Biomedical Center, Faculty of Medicine, LMU Munich, 82152, Planegg-Martinsried, Germany
| | - Kristian Schütze
- Institute of Immunology, University Medical Center Mainz, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Anna Seichter
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Hartmannstraße 14, D-91052, Erlangen, Germany
| | - Kristin Seré
- Institute for Biomedical Engineering, Department of Cell Biology, RWTH Aachen University Medical School, Aachen, Germany
- Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Athanasios Seretis
- Institute for Biomedical Aging Research, University of Innsbruck, Innsbruck, Austria
| | - Sieghart Sopper
- Internal Medicine V, Hematology and Oncology, Medical University of Innsbruck, Innsbruck, Austria
- Tyrolean Cancer Research Center, Innsbruck, Austria
| | - Helen Strandt
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Martina M Sykora
- Internal Medicine V, Hematology and Oncology, Medical University of Innsbruck, Innsbruck, Austria
- Tyrolean Cancer Research Center, Innsbruck, Austria
| | - Hannah Theobald
- Quantitative Systems Biology, Life and Medical Sciences (LIMES) Institute, University of Bonn, Germany
| | - Christoph H Tripp
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus (GRCC), U1015 INSERM, University Paris Saclay, Villejuif, France
| |
Collapse
|
28
|
Wu Z, Shih B, Macdonald J, Meunier D, Hogan K, Chintoan-Uta C, Gilhooley H, Hu T, Beltran M, Henderson NC, Sang HM, Stevens MP, McGrew MJ, Balic A. Development and function of chicken XCR1 + conventional dendritic cells. Front Immunol 2023; 14:1273661. [PMID: 37954617 PMCID: PMC10634274 DOI: 10.3389/fimmu.2023.1273661] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 10/06/2023] [Indexed: 11/14/2023] Open
Abstract
Conventional dendritic cells (cDCs) are antigen-presenting cells (APCs) that play a central role in linking innate and adaptive immunity. cDCs have been well described in a number of different mammalian species, but remain poorly characterised in the chicken. In this study, we use previously described chicken cDC specific reagents, a novel gene-edited chicken line and single-cell RNA sequencing (scRNAseq) to characterise chicken splenic cDCs. In contrast to mammals, scRNAseq analysis indicates that the chicken spleen contains a single, chemokine receptor XCR1 expressing, cDC subset. By sexual maturity the XCR1+ cDC population is the most abundant mononuclear phagocyte cell subset in the chicken spleen. scRNAseq analysis revealed substantial heterogeneity within the chicken splenic XCR1+ cDC population. Immature MHC class II (MHCII)LOW XCR1+ cDCs expressed a range of viral resistance genes. Maturation to MHCIIHIGH XCR1+ cDCs was associated with reduced expression of anti-viral gene expression and increased expression of genes related to antigen presentation via the MHCII and cross-presentation pathways. To visualise and transiently ablate chicken XCR1+ cDCs in situ, we generated XCR1-iCaspase9-RFP chickens using a CRISPR-Cas9 knockin transgenesis approach to precisely edit the XCR1 locus, replacing the XCR1 coding region with genes for a fluorescent protein (TagRFP), and inducible Caspase 9. After inducible ablation, the chicken spleen is initially repopulated by immature CD1.1+ XCR1+ cDCs. XCR1+ cDCs are abundant in the splenic red pulp, in close association with CD8+ T-cells. Knockout of XCR1 prevented this clustering of cDCs with CD8+ T-cells. Taken together these data indicate a conserved role for chicken and mammalian XCR1+ cDCs in driving CD8+ T-cells responses.
Collapse
Affiliation(s)
- Zhiguang Wu
- The Roslin Institute, University of Edinburgh, Midlothian, United Kingdom
| | - Barbara Shih
- The Roslin Institute, University of Edinburgh, Midlothian, United Kingdom
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster, United Kingdom
| | - Joni Macdonald
- The Roslin Institute, University of Edinburgh, Midlothian, United Kingdom
| | - Dominique Meunier
- The Roslin Institute, University of Edinburgh, Midlothian, United Kingdom
| | - Kris Hogan
- The Roslin Institute, University of Edinburgh, Midlothian, United Kingdom
| | | | - Hazel Gilhooley
- The Roslin Institute, University of Edinburgh, Midlothian, United Kingdom
| | - Tuanjun Hu
- The Roslin Institute, University of Edinburgh, Midlothian, United Kingdom
| | - Mariana Beltran
- Centre for Inflammation Research, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Neil C. Henderson
- Centre for Inflammation Research, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- Medical Research Council (MRC) Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Helen M. Sang
- The Roslin Institute, University of Edinburgh, Midlothian, United Kingdom
| | - Mark P. Stevens
- The Roslin Institute, University of Edinburgh, Midlothian, United Kingdom
| | - Michael J. McGrew
- The Roslin Institute, University of Edinburgh, Midlothian, United Kingdom
| | - Adam Balic
- The Roslin Institute, University of Edinburgh, Midlothian, United Kingdom
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
29
|
Okojie AK, Uweru JO, Coburn MA, Li S, Cao-Dao VD, Eyo UB. Distinguishing the effects of systemic CSF1R inhibition by PLX3397 on microglia and peripheral immune cells. J Neuroinflammation 2023; 20:242. [PMID: 37865779 PMCID: PMC10590528 DOI: 10.1186/s12974-023-02924-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 10/10/2023] [Indexed: 10/23/2023] Open
Abstract
Microglia, the primary immune cells of the central nervous system (CNS), are derived from the yolk sac and populate the brain during development. Once microglia migrate to the CNS, they are self-renewing and require CSF1R signaling for their maintenance. Pexidartinib (PLX3397, PLX), a small molecule inhibitor of the CSF1R, has been shown to effectively deplete microglia since microglial maintenance is CSF1R-dependent. There have, however, been several conflicting reports that have shown the potential off-target effects of PLX on peripheral immune cells particularly those of lymphoid origin. Given this controversy in the use of the PLX family of drugs, it has become important to ascertain to what extent PLX affects the peripheral immune profile in lymphoid (spleen, and bone marrow) and non-lymphoid (kidney, lungs, and heart) organs. PLX3397 chow treatment at 660 mg/kg for 7 days significantly reduced CD45+ macrophages, CX3CR1-GFP cells, CD11b+CD45intermediate cells, and P2RY12 expression in the brain. However, there were minimal effects on peripheral immune cells from both lymphoid and non-lymphoid organs except in the heart where there was a significant decrease in CD3+ cells, inflammatory and patrolling monocytes, and CD11b+Ly6G+ neutrophils. We then stimulated the immune system with 1 mg/kg of LPS which resulted in a significant reduction in the number of innate immune cells. In this context, PLX did not alter the cytokine profile in the serum and the brain of naïve mice but did so in the LPS-stimulated group resulting in a significant reduction in TNFα, IL-1α, IFN-γ and IL-1β. Furthermore, PLX did not alter locomotor activity in the open field test suggesting that microglia do not contribute to LPS-induced sickness behavior. Our results provide an assessment of immune cell populations with PLX3397 treatment on brain, lymphoid and non-lymphoid organs without and during LPS treatment that can serve as a resource for understanding consequences of such approaches.
Collapse
Affiliation(s)
- Akhabue K Okojie
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA.
- Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, VA, USA.
| | - Joseph O Uweru
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
- Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Morgan A Coburn
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
- Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Sihan Li
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Vivian D Cao-Dao
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA
- Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Ukpong B Eyo
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, USA.
- Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, VA, USA.
| |
Collapse
|
30
|
Blanco T, Singh RB, Nakagawa H, Taketani Y, Dohlman TH, Chen Y, Chauhan SK, Yin J, Dana R. Conventional type I migratory CD103 + dendritic cells are required for corneal allograft survival. Mucosal Immunol 2023; 16:711-726. [PMID: 36642378 PMCID: PMC10413378 DOI: 10.1016/j.mucimm.2022.12.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 12/14/2022] [Indexed: 01/15/2023]
Abstract
Corneal transplant rejection primarily occurs because of the T helper 1 (Th1) effector cell-mediated immune response of the host towards allogeneic tissue. The evidence suggests that type 1 migratory conventional CD103+ dendritic cells (CD103+DC1) acquire an immunosuppressive phenotype in the tumor environment; however, the involvement of CD103+DC1 in allograft survival continues to be an elusive question of great clinical significance in tissue transplantation. In this study, we assess the role of CD103+DC1 in suppressing Th1 alloreactivity against transplanted corneal allografts. The immunosuppressive function of CD103+DC1 has been extensively studied in non-transplantation settings. We found that host CD103+DC1 infiltrates the corneal graft and migrates to the draining lymph nodes to suppress alloreactive CD4+ Th1 cells via the programmed death-ligand 1 axis. The systemic depletion of CD103+ DC1 in allograft recipients leads to amplified Th1 activation, impaired Treg function, and increased rate of allograft rejection. Although allograft recipient Rag1 null mice reconstituted with naïve CD4+CD25- T cells efficiently generated peripheral Treg cells (pTreg), the CD103+DC1-depleted mice failed to generate pTreg. Furthermore, adoptive transfer of pTreg failed to rescue allografts in CD103+DC1-depleted recipients from rejection. These data demonstrate the critical role of CD103+DC1 in regulating host alloimmune responses.
Collapse
Affiliation(s)
- Tomas Blanco
- Laboratory of Corneal Immunology, Transplantation, and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, USA
| | - Rohan Bir Singh
- Laboratory of Corneal Immunology, Transplantation, and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, USA
| | - Hayate Nakagawa
- Laboratory of Corneal Immunology, Transplantation, and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, USA
| | - Yukako Taketani
- Laboratory of Corneal Immunology, Transplantation, and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, USA
| | - Thomas H Dohlman
- Laboratory of Corneal Immunology, Transplantation, and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, USA
| | - Yihe Chen
- Laboratory of Corneal Immunology, Transplantation, and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, USA
| | - Sunil K Chauhan
- Laboratory of Corneal Immunology, Transplantation, and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, USA
| | - Jia Yin
- Laboratory of Corneal Immunology, Transplantation, and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, USA
| | - Reza Dana
- Laboratory of Corneal Immunology, Transplantation, and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, USA.
| |
Collapse
|
31
|
Pereira da Costa M, Minutti CM, Piot C, Giampazolias E, Cardoso A, Cabeza-Cabrerizo M, Rogers NC, Lebrusant-Fernandez M, Iliakis CS, Wack A, Reis e Sousa C. Interplay between CXCR4 and CCR2 regulates bone marrow exit of dendritic cell progenitors. Cell Rep 2023; 42:112881. [PMID: 37523265 DOI: 10.1016/j.celrep.2023.112881] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 05/02/2023] [Accepted: 07/13/2023] [Indexed: 08/02/2023] Open
Abstract
Conventional dendritic cells (cDCs) are found in most tissues and play a key role in initiation of immunity. cDCs require constant replenishment from progenitors called pre-cDCs that develop in the bone marrow (BM) and enter the blood circulation to seed all tissues. This process can be markedly accelerated in response to inflammation (emergency cDCpoiesis). Here, we identify two populations of BM pre-cDC marked by differential expression of CXCR4. We show that CXCR4lo cells constitute the migratory pool of BM pre-cDCs, which exits the BM and can be rapidly mobilized during challenge. We further show that exit of CXCR4lo pre-cDCs from BM at steady state is partially dependent on CCR2 and that CCR2 upregulation in response to type I IFN receptor signaling markedly increases efflux during infection with influenza A virus. Our results highlight a fine balance between retention and efflux chemokine cues that regulates steady-state and emergency cDCpoiesis.
Collapse
Affiliation(s)
| | - Carlos M Minutti
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Cécile Piot
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Evangelos Giampazolias
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Ana Cardoso
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Mar Cabeza-Cabrerizo
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Neil C Rogers
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Marta Lebrusant-Fernandez
- Immune Responses to Lipids Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Chrysante S Iliakis
- Immunoregulation Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Andreas Wack
- Immunoregulation Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Caetano Reis e Sousa
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.
| |
Collapse
|
32
|
Silva–Sanchez A, Meza–Perez S, Liu M, Stone SL, Flores–Romo L, Ubil E, Lund FE, Rosenberg AF, Randall TD. Activation of regulatory dendritic cells by Mertk coincides with a temporal wave of apoptosis in neonatal lungs. Sci Immunol 2023; 8:eadc9081. [PMID: 37327322 PMCID: PMC10351240 DOI: 10.1126/sciimmunol.adc9081] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 05/24/2023] [Indexed: 06/18/2023]
Abstract
Multiple mechanisms restrain inflammation in neonates, most likely to prevent tissue damage caused by overly robust immune responses against newly encountered pathogens. Here, we identify a population of pulmonary dendritic cells (DCs) that express intermediate levels of CD103 (CD103int) and appear in the lungs and lung-draining lymph nodes of mice between birth and 2 weeks of age. CD103int DCs express XCR1 and CD205 and require expression of the transcription factor BATF3 for development, suggesting that they belong to the cDC1 lineage. In addition, CD103int DCs express CCR7 constitutively and spontaneously migrate to the lung-draining lymph node, where they promote stromal cell maturation and lymph node expansion. CD103int DCs mature independently of microbial exposure and TRIF- or MyD88-dependent signaling and are transcriptionally related to efferocytic and tolerogenic DCs as well as mature, regulatory DCs. Correlating with this, CD103int DCs show limited ability to stimulate proliferation and IFN-γ production by CD8+ T cells. Moreover, CD103int DCs acquire apoptotic cells efficiently, in a process that is dependent on the expression of the TAM receptor, Mertk, which drives their homeostatic maturation. The appearance of CD103int DCs coincides with a temporal wave of apoptosis in developing lungs and explains, in part, dampened pulmonary immunity in neonatal mice. Together, these data suggest a mechanism by which DCs sense apoptotic cells at sites of noninflammatory tissue remodeling, such as tumors or the developing lungs, and limit local T cell responses.
Collapse
Affiliation(s)
- Aaron Silva–Sanchez
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL
| | - Selene Meza–Perez
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL
| | - Mingyong Liu
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL
| | - Sara L Stone
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL
| | - Leopoldo Flores–Romo
- Department of Cell Biology, Center for Advanced Research, The National Polytechnic Institute, Cinvestav–IPN, Mexico City, Mexico
| | - Eric Ubil
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL
| | - Frances E. Lund
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL
| | | | - Troy D. Randall
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
33
|
Yamashita H, Matsuhara H, Tanaka H, Inagaki N, Tsutsui M. Oral allergy induction through skin exposure to previously tolerated food antigens in murine models. J Pharmacol Sci 2023; 152:76-85. [PMID: 37169482 DOI: 10.1016/j.jphs.2023.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/16/2023] [Accepted: 03/27/2023] [Indexed: 05/13/2023] Open
Abstract
Food allergies (FAs) are caused by a failure of the immune system to regulate oral tolerance (OT). The use of soap containing hydrolyzed wheat overrides acquired OT to wheat through skin exposure. However, in mouse models, the experimental OT is robust, suggesting that acquired OT to allergens prevents the development of FAs. We aimed to analyze the mechanisms and developed a mouse model of FA that overrides acquired OT via skin exposure. Three murine FA models (intraperitoneal [IP], epicutaneous [EC], and intradermal [ID]) were compared to evaluate if allergies to ovalbumin (OVA) that had been previously tolerated orally could be induced. In the ID model, OT was overridden, and allergic reactions of severe anaphylaxis were developed. To analyze this effect in the ID model, we measured the migration of dendritic cells (DCs) into lymph nodes. The induction of OT promoted the migration of CD103+ dermal DCs; moreover, repeated percutaneous doses of OVA for sensitization gradually increased the migration of CD11b+ dermal DCs. The difference in the proportion of regulatory T cells between ID-sensitized groups at the first ID injection disappeared at the tenth injection. Although OT was robust in the IP model, ID sensitization was found to override OT.
Collapse
Affiliation(s)
- Hirotaka Yamashita
- Department of Pharmacology, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Okinawa, 903-0215, Japan.
| | - Hiroki Matsuhara
- Immuno-Pharmacology, Field of Biofunctional Control, Medical Information Science Division, United Graduate School of Drug Discovery and Medical Information Science, Gifu University, 1-1 Yanaido, Gifu 501-1194, Japan
| | - Hiroyuki Tanaka
- Immuno-Pharmacology, Field of Biofunctional Control, Medical Information Science Division, United Graduate School of Drug Discovery and Medical Information Science, Gifu University, 1-1 Yanaido, Gifu 501-1194, Japan; Laboratory of Immunobiology, Department of Biofunctional Analysis, Gifu Pharmaceutical University, 1-25-4 Daigakunishi, Gifu 501-1196, Japan
| | - Naoki Inagaki
- Department of Pharmacy, Faculty of Pharmacy, Gifu University of Medical Science, 4-3-3 Nijigaoka, Kani City, Gifu 509-0293, Japan
| | - Masato Tsutsui
- Department of Pharmacology, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Okinawa, 903-0215, Japan
| |
Collapse
|
34
|
Chen T, Cao Q, Wang R, Zheng G, Azmi F, Lee VW, Wang YM, Li H, Yu D, Rogers NM, Alexander SI, Harris DCH, Wang Y. Attenuation of renal injury by depleting cDC1 and by repurposing Flt3 inhibitor in anti-GBM disease. Clin Immunol 2023; 250:109295. [PMID: 36933629 DOI: 10.1016/j.clim.2023.109295] [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: 11/20/2022] [Accepted: 03/15/2023] [Indexed: 03/18/2023]
Abstract
Previous studies found cDC1s to be protective in early stage anti-GBM disease through Tregs, but pathogenic in late stage Adriamycin nephropathy through CD8+ T cells. Flt3 ligand is a growth factor essential for cDC1 development and Flt3 inhibitors are currently used for cancer treatment. We conducted this study to clarify the role and mechanisms of effects of cDC1s at different time points in anti-GBM disease. In addition, we aimed to utilize drug repurposing of Flt3 inhibitors to target cDC1s as a treatment of anti-GBM disease. We found that in human anti-GBM disease, the number of cDC1s increased significantly, proportionally more than cDC2s. The number of CD8+ T cells also increased significantly and their number correlated with cDC1 number. In XCR1-DTR mice, late (day 12-21) but not early (day 3-12) depletion of cDC1s attenuated kidney injury in mice with anti-GBM disease. cDC1s separated from kidneys of anti-GBM disease mice were found to have a pro-inflammatory phenotype (i.e. express high level of IL-6, IL-12 and IL-23) in late but not early stage. In the late depletion model, the number of CD8+ T cells was also reduced, but not Tregs. CD8+ T cells separated from kidneys of anti-GBM disease mice expressed high levels of cytotoxic molecules (granzyme B and perforin) and inflammatory cytokines (TNF-α and IFN-γ), and their expression reduced significantly after cDC1 depletion with diphtheria toxin. These findings were reproduced using a Flt3 inhibitor in wild type mice. Therefore, cDC1s are pathogenic in anti-GBM disease through activation of CD8+ T cells. Flt3 inhibition successfully attenuated kidney injury through depletion of cDC1s. Repurposing Flt3 inhibitors has potential as a novel therapeutic strategy for anti-GBM disease.
Collapse
Affiliation(s)
- Titi Chen
- The University of Sydney, Camperdown, NSW 2006, Australia; The Westmead Institute for Medical Research, Hawkesbury Road, Westmead, NSW 2145, Australia; Department of Renal Medicine, Westmead Hospital, Hawkesbury Road, Westmead, NSW 2145, Australia.
| | - Qi Cao
- The University of Sydney, Camperdown, NSW 2006, Australia; The Westmead Institute for Medical Research, Hawkesbury Road, Westmead, NSW 2145, Australia
| | - Ruifeng Wang
- The University of Sydney, Camperdown, NSW 2006, Australia; The Westmead Institute for Medical Research, Hawkesbury Road, Westmead, NSW 2145, Australia; Department of Nephrology, The Second Hospital of Anhui Medical University, Anhui 230000, China
| | - Guoping Zheng
- The University of Sydney, Camperdown, NSW 2006, Australia; The Westmead Institute for Medical Research, Hawkesbury Road, Westmead, NSW 2145, Australia
| | - Farhana Azmi
- The University of Sydney, Camperdown, NSW 2006, Australia; The Westmead Institute for Medical Research, Hawkesbury Road, Westmead, NSW 2145, Australia
| | - Vincent W Lee
- The University of Sydney, Camperdown, NSW 2006, Australia; The Westmead Institute for Medical Research, Hawkesbury Road, Westmead, NSW 2145, Australia; Department of Renal Medicine, Westmead Hospital, Hawkesbury Road, Westmead, NSW 2145, Australia
| | - Yuan Ming Wang
- Centre for Kidney Research, Children's Hospital at Westmead, Sydney, NSW 2145, Australia
| | - Hongqi Li
- The University of Sydney, Camperdown, NSW 2006, Australia; The Westmead Institute for Medical Research, Hawkesbury Road, Westmead, NSW 2145, Australia; The Department of Gerontology, Anhui Provincial Hospital, the first affiliated Hospital of University of Science and Technology of China, Hefei 230001, China
| | - Di Yu
- Faculty of Medicine, The University of Queensland Diamantina Institute, St Lucia, QLD 4072, Australia
| | - Natasha M Rogers
- The University of Sydney, Camperdown, NSW 2006, Australia; The Westmead Institute for Medical Research, Hawkesbury Road, Westmead, NSW 2145, Australia; Department of Renal Medicine, Westmead Hospital, Hawkesbury Road, Westmead, NSW 2145, Australia
| | - Stephen I Alexander
- Centre for Kidney Research, Children's Hospital at Westmead, Sydney, NSW 2145, Australia
| | - David C H Harris
- The University of Sydney, Camperdown, NSW 2006, Australia; The Westmead Institute for Medical Research, Hawkesbury Road, Westmead, NSW 2145, Australia; Department of Renal Medicine, Westmead Hospital, Hawkesbury Road, Westmead, NSW 2145, Australia
| | - Yiping Wang
- The University of Sydney, Camperdown, NSW 2006, Australia; The Westmead Institute for Medical Research, Hawkesbury Road, Westmead, NSW 2145, Australia
| |
Collapse
|
35
|
Tsai J, Kaneko K, Suh AJ, Bockman R, Park-Min KH. Origin of Osteoclasts: Osteoclast Precursor Cells. J Bone Metab 2023; 30:127-140. [PMID: 37449346 PMCID: PMC10346003 DOI: 10.11005/jbm.2023.30.2.127] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/27/2023] [Accepted: 04/27/2023] [Indexed: 07/18/2023] Open
Abstract
Osteoclasts are multinucleated bone-resorbing cells and a key player in bone remodeling for health and disease. Since the discovery of osteoclasts in 1873, the structure and function of osteoclasts and the molecular and cellular mechanisms of osteoclastogenesis have been extensively studied. Moreover, it has been well established that osteoclasts are differentiated in vitro from myeloid cells such as bone marrow macrophages or monocytes. The concept showing that osteoclasts are derived from a specific population (named osteoclast precursor cells [OCPs]) among myeloid cells has been long hypothesized. However, the specific precursor population of osteoclasts is not clearly defined yet. A growing body of work provides evidence of the developmental origin and lifespan of murine osteoclasts, particularly in vivo. Here, we review the emerging evidence that supports the existence of OCPs and discuss current insights into their identity.
Collapse
Affiliation(s)
- Jefferson Tsai
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY,
USA
| | - Kaichi Kaneko
- Division of Rheumatology, Department of Internal Medicine, Toho University Sakura Medical Center, Chiba,
Japan
| | - Andrew J. Suh
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY,
USA
| | - Richard Bockman
- Division of Endocrinology and Metabolism, Hospital for Special Surgery, New York, NY,
USA
- Department of Medicine, Weill Cornell Medical College, New York, NY,
USA
| | - Kyung-Hyun Park-Min
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY,
USA
- Department of Medicine, Weill Cornell Medical College, New York, NY,
USA
- BCMB Allied Program, Weill Cornell Graduate School of Medical Sciences, New York, NY,
USA
| |
Collapse
|
36
|
Bhargava R, Li H, Tsokos GC. Pathogenesis of lupus nephritis: the contribution of immune and kidney resident cells. Curr Opin Rheumatol 2023; 35:107-116. [PMID: 35797522 DOI: 10.1097/bor.0000000000000887] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
PURPOSE OF REVIEW Lupus nephritis is associated with significant mortality and morbidity. We lack effective therapeutics and biomarkers mostly because of our limited understanding of its complex pathogenesis. We aim to present an overview of the recent advances in the field to gain a deeper understanding of the underlying cellular and molecular mechanisms involved in lupus nephritis pathogenesis. RECENT FINDINGS Recent studies have identified distinct roles for each resident kidney cell in the pathogenesis of lupus nephritis. Podocytes share many elements of innate and adaptive immune cells and they can present antigens and participate in the formation of crescents in coordination with parietal epithelial cells. Mesangial cells produce pro-inflammatory cytokines and secrete extracellular matrix contributing to glomerular fibrosis. Tubular epithelial cells modulate the milieu of the interstitium to promote T cell infiltration and formation of tertiary lymphoid organs. Modulation of specific genes in kidney resident cells can ward off the effectors of the autoimmune response including autoantibodies, cytokines and immune cells. SUMMARY The development of lupus nephritis is multifactorial involving genetic susceptibility, environmental triggers and systemic inflammation. However, the role of resident kidney cells in the development of lupus nephritis is becoming more defined and distinct. More recent studies point to the restoration of kidney resident cell function using cell targeted approaches to prevent and treat lupus nephritis.
Collapse
Affiliation(s)
- Rhea Bhargava
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard, Medical School, Boston, Massachusetts, USA
| | | | | |
Collapse
|
37
|
MHC-dressing on dendritic cells: Boosting anti-tumor immunity via unconventional tumor antigen presentation. Semin Immunol 2023; 66:101710. [PMID: 36640616 DOI: 10.1016/j.smim.2023.101710] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 12/21/2022] [Accepted: 01/07/2023] [Indexed: 01/15/2023]
Abstract
Dendritic cells are crucial for anti-tumor immune responses due to their ability to activate cytotoxic effector CD8+ T cells. Canonically, in anti-tumor immunity, dendritic cells activate CD8+ T cells in a process termed cross-presentation. Recent studies have demonstrated that another type of antigen presentation, MHC-dressing, also serves to activate CD8+ T cells against tumor cell-derived antigens. Understanding MHC-dressing's specific contributions to anti-tumor immunity can open up novel therapeutic avenues. In this review, we summarize the early studies that identified MHC-dressing as a relevant antigen presentation pathway before diving into a deeper discussion of the biology of MHC-dressing, focusing in particular on which dendritic cell subsets are most capable of performing MHC-dressing and how MHC-dressing compares to other forms of antigen presentation. We conclude by discussing the implications MHC-dressing has for anti-tumor immunity.
Collapse
|
38
|
Backer RA, Probst HC, Clausen BE. Classical DC2 subsets and monocyte-derived DC: Delineating the developmental and functional relationship. Eur J Immunol 2023; 53:e2149548. [PMID: 36642930 DOI: 10.1002/eji.202149548] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 11/08/2023] [Accepted: 01/13/2023] [Indexed: 01/17/2023]
Abstract
To specifically tailor immune responses to a given pathogenic threat, dendritic cells (DC) are highly heterogeneous and comprise many specialized subtypes, including conventional DC (cDC) and monocyte-derived DC (MoDC), each with distinct developmental and functional characteristics. However, the functional relationship between cDC and MoDC is not fully understood, as the overlapping phenotypes of certain type 2 cDC (cDC2) subsets and MoDC do not allow satisfactory distinction of these cells in the tissue, particularly during inflammation. However, precise cDC2 and MoDC classification is required for studies addressing how these diverse cell types control immune responses and is therefore currently one of the major interests in the field of cDC research. This review will revise murine cDC2 and MoDC biology in the steady state and under inflammatory conditions and discusses the commonalities and differences between ESAMlo cDC2, inflammatory cDC2, and MoDC and their relative contribution to the initiation, propagation, and regulation of immune responses.
Collapse
Affiliation(s)
- Ronald A Backer
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Hans Christian Probst
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Institute for Immunology, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Björn E Clausen
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| |
Collapse
|
39
|
Watanabe T, Lam C, Oliver J, Oishi H, Teskey G, Beber S, Boonstra K, Mauricio Umaña J, Buhari H, Joe B, Guan Z, Horie M, Keshavjee S, Martinu T, Juvet SC. Donor Batf3 inhibits murine lung allograft rejection and airway fibrosis. Mucosal Immunol 2023; 16:104-120. [PMID: 36842540 DOI: 10.1016/j.mucimm.2023.02.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 02/14/2023] [Indexed: 02/28/2023]
Abstract
Chronic lung allograft dysfunction (CLAD) limits survival after lung transplantation. Noxious stimuli entering the airways foster CLAD development. Classical dendritic cells (cDCs) link innate and adaptive immunity and exhibit regional and functional specialization in the lung. The transcription factor basic leucine zipper ATF-like 3 (BATF3) is absolutely required for the development of type 1 cDCs (cDC1s), which reside in the airway epithelium and have variable responses depending on the context. We studied the role of BATF3 in a mouse minor alloantigen-mismatched orthotopic lung transplant model of CLAD with and without airway inflammation triggered by repeated administration of intratracheal lipopolysaccharide (LPS). We found that cDC1s accumulated in allografts compared with isografts and that donor cDC1s were gradually replaced by recipient cDC1s. LPS administration increased the number of cDC1s and enhanced their state of activation. We found that Batf3-/- recipient mice experienced reduced acute rejection in response to LPS; in contrast, Batf3-/- donor grafts underwent enhanced lung and skin allograft rejection and drove augmented recipient cluster of differentiation 8+ T-cell expansion in the absence of LPS. Our findings suggest that donor and recipient cDC1s have differing and context-dependent roles and may represent a therapeutic target in lung transplantation.
Collapse
Affiliation(s)
- Tatsuaki Watanabe
- Latner Thoracic Research Laboratories, University Health Network, University of Toronto, Toronto, Canada; Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Christina Lam
- Latner Thoracic Research Laboratories, University Health Network, University of Toronto, Toronto, Canada
| | - Jillian Oliver
- Latner Thoracic Research Laboratories, University Health Network, University of Toronto, Toronto, Canada
| | - Hisashi Oishi
- Latner Thoracic Research Laboratories, University Health Network, University of Toronto, Toronto, Canada; Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Grace Teskey
- Latner Thoracic Research Laboratories, University Health Network, University of Toronto, Toronto, Canada
| | - Samuel Beber
- Latner Thoracic Research Laboratories, University Health Network, University of Toronto, Toronto, Canada
| | - Kristen Boonstra
- Latner Thoracic Research Laboratories, University Health Network, University of Toronto, Toronto, Canada
| | - Juan Mauricio Umaña
- Latner Thoracic Research Laboratories, University Health Network, University of Toronto, Toronto, Canada
| | - Hifza Buhari
- Latner Thoracic Research Laboratories, University Health Network, University of Toronto, Toronto, Canada
| | - Betty Joe
- Latner Thoracic Research Laboratories, University Health Network, University of Toronto, Toronto, Canada
| | - Zehong Guan
- Latner Thoracic Research Laboratories, University Health Network, University of Toronto, Toronto, Canada
| | - Miho Horie
- Joint Department of Medical Imaging, University Health Network, Toronto, Canada
| | - Shaf Keshavjee
- Latner Thoracic Research Laboratories, University Health Network, University of Toronto, Toronto, Canada
| | - Tereza Martinu
- Latner Thoracic Research Laboratories, University Health Network, University of Toronto, Toronto, Canada
| | - Stephen C Juvet
- Latner Thoracic Research Laboratories, University Health Network, University of Toronto, Toronto, Canada.
| |
Collapse
|
40
|
Erra Diaz F, Mazzitelli I, Bleichmar L, Melucci C, Thibodeau A, Dalotto Moreno T, Marches R, Rabinovich GA, Ucar D, Geffner J. Concomitant inhibition of PPARγ and mTORC1 induces the differentiation of human monocytes into highly immunogenic dendritic cells. Cell Rep 2023; 42:112156. [PMID: 36842088 DOI: 10.1016/j.celrep.2023.112156] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/29/2022] [Accepted: 02/08/2023] [Indexed: 02/27/2023] Open
Abstract
Monocytes can differentiate into macrophages (Mo-Macs) or dendritic cells (Mo-DCs). The cytokine granulocyte-macrophage colony-stimulating factor (GM-CSF) induces the differentiation of monocytes into Mo-Macs, while the combination of GM-CSF/interleukin (IL)-4 is widely used to generate Mo-DCs for clinical applications and to study human DC biology. Here, we report that pharmacological inhibition of the nuclear receptor peroxisome proliferator-activated receptor gamma (PPARγ) in the presence of GM-CSF and the absence of IL-4 induces monocyte differentiation into Mo-DCs. Remarkably, we find that simultaneous inhibition of PPARγ and the nutrient sensor mammalian target of rapamycin complex 1 (mTORC1) induces the differentiation of Mo-DCs with stronger phenotypic stability, superior immunogenicity, and a transcriptional profile characterized by a strong type I interferon (IFN) signature, a lower expression of a large set of tolerogenic genes, and the differential expression of several transcription factors compared with GM-CSF/IL-4 Mo-DCs. Our findings uncover a pathway that tailors Mo-DC differentiation with potential implications in the fields of DC vaccination and cancer immunotherapy.
Collapse
Affiliation(s)
- Fernando Erra Diaz
- Facultad de Medicina, Instituto de Investigaciones Biomédicas en Retrovirus y SIDA, Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina
| | - Ignacio Mazzitelli
- Facultad de Medicina, Instituto de Investigaciones Biomédicas en Retrovirus y SIDA, Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina
| | - Lucía Bleichmar
- Facultad de Medicina, Instituto de Investigaciones Biomédicas en Retrovirus y SIDA, Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina
| | - Claudia Melucci
- Facultad de Medicina, Instituto de Investigaciones Biomédicas en Retrovirus y SIDA, Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina
| | - Asa Thibodeau
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Tomás Dalotto Moreno
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - Radu Marches
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Gabriel A Rabinovich
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina; Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Duygu Ucar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA; Institute for Systems Genomics, University of Connecticut Health Center, Farmington, CT 06030, USA.
| | - Jorge Geffner
- Facultad de Medicina, Instituto de Investigaciones Biomédicas en Retrovirus y SIDA, Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina.
| |
Collapse
|
41
|
Cui TX, Brady AE, Zhang YJ, Fulton CT, Goldsmith AM, Popova AP. Early-life hyperoxia-induced Flt3L drives neonatal lung dendritic cell expansion and proinflammatory responses. Front Immunol 2023; 14:1116675. [PMID: 36845082 PMCID: PMC9950736 DOI: 10.3389/fimmu.2023.1116675] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 01/26/2023] [Indexed: 02/12/2023] Open
Abstract
Premature infants with chronic lung disease, bronchopulmonary dysplasia (BPD), develop recurrent cough and wheezing following respiratory viral infections. The mechanisms driving the chronic respiratory symptoms are ill-defined. We have shown that hyperoxic exposure of neonatal mice (a model of BPD) increases the activated lung CD103+ dendritic cells (DCs) and these DCs are required for exaggerated proinflammatory responses to rhinovirus (RV) infection. Since CD103+ DC are essential for specific antiviral responses and their development depends on the growth factor Flt3L, we hypothesized that early-life hyperoxia stimulates Flt3L expression leading to expansion and activation of lung CD103+ DCs and this mediates inflammation. We found that hyperoxia numerically increased and induced proinflammatory transcriptional signatures in neonatal lung CD103+ DCs, as well as CD11bhi DCs. Hyperoxia also increased Flt3L expression. Anti-Flt3L antibody blocked CD103+ DC development in normoxic and hyperoxic conditions, and while it did not affect the baseline number of CD11bhi DCs, it neutralized the effect of hyperoxia on these cells. Anti-Flt3L also inhibited hyperoxia-induced proinflammatory responses to RV. In tracheal aspirates from preterm infants mechanically-ventilated for respiratory distress in the first week of life levels of FLT3L, IL-12p40, IL-12p70 and IFN-γ were higher in infants who went on to develop BPD and FLT3L levels positively correlated with proinflammatory cytokines levels. This work highlights the priming effect of early-life hyperoxia on lung DC development and function and the contribution of Flt3L in driving these effects.
Collapse
Affiliation(s)
| | | | | | | | | | - Antonia P. Popova
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
42
|
Redondo-Urzainqui A, Hernández-García E, Cook ECL, Iborra S. Dendritic cells in energy balance regulation. Immunol Lett 2023; 253:19-27. [PMID: 36586424 DOI: 10.1016/j.imlet.2022.12.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/21/2022] [Accepted: 12/26/2022] [Indexed: 12/30/2022]
Abstract
Besides their well-known role in initiating adaptive immune responses, several groups have studied the role of dendritic cells (DCs) in the context of chronic metabolic inflammation, such as in diet-induced obesity (DIO) or metabolic-associated fatty liver disease. DCs also have an important function in maintaining metabolic tissue homeostasis in steady-state conditions. In this review, we will briefly describe the different DC subsets, the murine models available to assess their function, and discuss the role of DCs in regulating energy balance and maintaining tissue homeostasis.
Collapse
Affiliation(s)
- Ana Redondo-Urzainqui
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, 28040, Spain
| | - Elena Hernández-García
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, 28040, Spain
| | - Emma Clare Laura Cook
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, 28040, Spain.
| | - Salvador Iborra
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, 28040, Spain.
| |
Collapse
|
43
|
Duluc D, Sisirak V. Origin, Phenotype, and Function of Mouse Dendritic Cell Subsets. Methods Mol Biol 2023; 2618:3-16. [PMID: 36905505 DOI: 10.1007/978-1-0716-2938-3_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Dendritic cells are cells of hematopoietic origin that are specialized in antigen presentation and instruction of innate and adaptive immune responses. They are a heterogenous group of cells populating lymphoid organs and most tissues. Dendritic cells are commonly separated in three main subsets that differ in their developmental paths, phenotype, and functions. Most studies on dendritic cells were done primarily in mice; therefore, in this chapter, we propose to summarize the current knowledge and recent progress on mouse dendritic cell subsets' development, phenotype, and functions.
Collapse
Affiliation(s)
- Dorothée Duluc
- Université de Bordeaux, CNRS, ImmunoConcEpT, UMR 5164, Bordeaux, France.
| | - Vanja Sisirak
- UMR CNRS 5164 - Immunoconcept, Université de Bordeaux, Bordeaux, France.
| |
Collapse
|
44
|
Plackoska V, Shaban D, Nijnik A. Hematologic dysfunction in cancer: Mechanisms, effects on antitumor immunity, and roles in disease progression. Front Immunol 2022; 13:1041010. [PMID: 36561751 PMCID: PMC9763314 DOI: 10.3389/fimmu.2022.1041010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 11/17/2022] [Indexed: 12/12/2022] Open
Abstract
With the major advances in cancer immunology and immunotherapy, it is critical to consider that most immune cells are short-lived and need to be continuously replenished from hematopoietic stem and progenitor cells. Hematologic abnormalities are prevalent in cancer patients, and many ground-breaking studies over the past decade provide insights into their underlying cellular and molecular mechanisms. Such studies demonstrate that the dysfunction of hematopoiesis is more than a side-effect of cancer pathology, but an important systemic feature of cancer disease. Here we review these many advances, covering the cancer-associated phenotypes of hematopoietic stem and progenitor cells, the dysfunction of myelopoiesis and erythropoiesis, the importance of extramedullary hematopoiesis in cancer disease, and the developmental origins of tumor associated macrophages. We address the roles of many secreted mediators, signaling pathways, and transcriptional and epigenetic mechanisms that mediate such hematopoietic dysfunction. Furthermore, we discuss the important contribution of the hematopoietic dysfunction to cancer immunosuppression, the possible avenues for therapeutic intervention, and highlight the unanswered questions and directions for future work. Overall, hematopoietic dysfunction is established as an active component of the cancer disease mechanisms and an important target for therapeutic intervention.
Collapse
Affiliation(s)
- Viktoria Plackoska
- Department of Physiology, McGill University, Montreal, QC, Canada,McGill University Research Centre on Complex Traits, McGill University, Montreal, QC, Canada
| | - Dania Shaban
- Department of Physiology, McGill University, Montreal, QC, Canada,McGill University Research Centre on Complex Traits, McGill University, Montreal, QC, Canada
| | - Anastasia Nijnik
- Department of Physiology, McGill University, Montreal, QC, Canada,McGill University Research Centre on Complex Traits, McGill University, Montreal, QC, Canada,*Correspondence: Anastasia Nijnik,
| |
Collapse
|
45
|
Deng N, Zuo X, lin Q, Wang T, Li Y, Zhong J, Ni H, Chen Q, Ding X, Yu H, Nie H. Low-dose 5-fluorouracil ameliorates Th2 responses through the induction of apoptotic cell death of lung monocyte-derived dendritic cells in asthma. Biomed Pharmacother 2022; 156:113875. [DOI: 10.1016/j.biopha.2022.113875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/26/2022] [Accepted: 10/13/2022] [Indexed: 11/24/2022] Open
|
46
|
Wolf G, Gerber AN, Fasana ZG, Rosenberg K, Singh NJ. Acute effects of FLT3L treatment on T cells in intact mice. Sci Rep 2022; 12:19487. [PMID: 36376544 PMCID: PMC9662129 DOI: 10.1038/s41598-022-24126-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
Peripheral T cells express a diverse repertoire of antigen-specific receptors, which together protect against the full range of pathogens. In this context, the total repertoire of memory T cells which are maintained by trophic signals, long after pathogen clearance, is critical. Since these trophic factors include cytokines and self-peptide-MHC, both of which are available from endogenous antigen-presenting cells (APC), we hypothesized that enhancing APC numbers in vivo can be a viable strategy to amplify the population of memory T cells. We evaluated this by acutely treating intact mice with FMS-like tyrosine kinase 3 ligand (Flt3l), which promotes expansion of APCs. Here we report that this treatment allowed for, an expansion of effector-memory CD4+ and CD8+ T cells as well as an increase in their expression of KLRG1 and CD25. In the lymph nodes and spleen, the expansion was limited to a specific CD8 (CD44-low but CD62L-) subset. Functionally, this subset is distinct from naïve T cells and could produce significant amounts of effector cytokines upon restimulation. Taken together, these data suggest that the administration of Flt3L can impact both APC turnover as well as a corresponding flux of specific subsets of CD8+ T cells in an intact peripheral immune compartment.
Collapse
Affiliation(s)
- Gideon Wolf
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W Baltimore St., HSF1, Room 380, Baltimore, MD, 21201, USA
| | - Allison N Gerber
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W Baltimore St., HSF1, Room 380, Baltimore, MD, 21201, USA
| | - Zachary G Fasana
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W Baltimore St., HSF1, Room 380, Baltimore, MD, 21201, USA
| | - Kenneth Rosenberg
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W Baltimore St., HSF1, Room 380, Baltimore, MD, 21201, USA
| | - Nevil J Singh
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W Baltimore St., HSF1, Room 380, Baltimore, MD, 21201, USA.
| |
Collapse
|
47
|
Delgado M, Lennon-Duménil AM. How cell migration helps immune sentinels. Front Cell Dev Biol 2022; 10:932472. [PMID: 36268510 PMCID: PMC9577558 DOI: 10.3389/fcell.2022.932472] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 09/13/2022] [Indexed: 12/01/2022] Open
Abstract
The immune system relies on the migratory capacity of its cellular components, which must be mobile in order to defend the host from invading micro-organisms or malignant cells. This applies in particular to immune sentinels from the myeloid lineage, i.e. macrophages and dendritic cells. Cell migration is already at work during mammalian early development, when myeloid cell precursors migrate from the yolk sac, an extra embryonic structure, to colonize tissues and form the pool of tissue-resident macrophages. Later, this is accompanied by a migration wave of precursors and monocytes from the bone marrow to secondary lymphoid organs and the peripheral tissues. They differentiate into DCs and monocyte-derived macrophages. During adult life, cell migration endows immune cells with the ability to patrol their environment as well as to circulate between peripheral tissues and lymphoid organs. Hence migration of immune cells is key to building an efficient defense system for an organism. In this review, we will describe how cell migratory capacity regulates the various stages in the life of myeloid cells from development to tissue patrolling, and migration to lymph nodes. We will focus on the role of the actin cytoskeletal machinery and its regulators, and how it contributes to the establishment and function of the immune system.
Collapse
|
48
|
Tsymbalyuk O, Gerzanich V, Simard JM, Rathinam CV. Traumatic brain injury alters dendritic cell differentiation and distribution in lymphoid and non-lymphoid organs. J Neuroinflammation 2022; 19:238. [PMID: 36183126 PMCID: PMC9526328 DOI: 10.1186/s12974-022-02609-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 09/24/2022] [Indexed: 11/10/2022] Open
Abstract
Background Pathophysiological consequences of traumatic brain injury (TBI) mediated secondary injury remain incompletely understood. In particular, the impact of TBI on the differentiation and maintenance of dendritic cells (DCs), which are regarded as the most professional antigen presenting cells of the immune system, remains completely unknown. Here, we report that DC-differentiation, maintenance and functions are altered on day 3 and day 7 after TBI. Methods Long bones, spleen, peripheral lymph nodes (pLNs), mesenteric lymph nodes (mLNs), liver, lungs, skin and blood were collected from mice with either moderate-level cortical impact (CCI) or sham on day 1, day 3 or day 7 after TBI. Bone marrow cells were isolated from the tibias and femurs of hind limb through flushing. Tissues were digested with Collagenase-D and DNase I. Skin biopsies were digested in the presence of liberase + DNase I. Single cell suspensions were made, red blood cells were lysed with Ammonium chloride (Stem Cell Technology) and subsequently filtered using a 70 μM nylon mesh. DC subsets of the tissues and DC progenitors of the BM were identified through 10-color flow cytometry-based immunophenotyping studies. Intracellular reactive oxygen species (ROS) were identified through H2DCFDA staining. Results Our studies identify that; (1) frequencies and absolute numbers of DCs in the spleen and BM are altered on day 3 and day 7 after TBI; (2) surface expression of key molecules involved in antigen presentation of DCs were affected on day 3 and day 7 after TBI; (3) distribution and functions of tissue-specific DC subsets of both circulatory and lymphatic systems were imbalanced following TBI; (4) early differentiation program of DCs, especially the commitment of hematopoietic stem cells to common DC progenitors (CDPs), were deregulated after TBI; and (5) intracellular ROS levels were reduced in DC progenitors and differentiated DCs on day 3 and day 7 after TBI. Conclusions Our data demonstrate, for the first time, that TBI affects the distribution pattern of DCs and induces an imbalance among DC subsets in both lymphoid and non-lymphoid organs. In addition, the current study demonstrates that TBI results in reduced levels of ROS in DCs on day 3 and day 7 after TBI, which may explain altered DC differentiation paradigm following TBI. A deeper understanding on the molecular mechanisms that contribute to DC defects following TBI would be essential and beneficial in treating infections in patients with acute central nervous system (CNS) injuries, such as TBI, stroke and spinal cord injury.
Collapse
Affiliation(s)
- Orest Tsymbalyuk
- Department of Neurosurgery, University of Maryland School of Medicine, MD, Baltimore, USA
| | - Volodymyr Gerzanich
- Department of Neurosurgery, University of Maryland School of Medicine, MD, Baltimore, USA
| | - J Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, MD, Baltimore, USA.,Research Service, Veterans Affairs Maryland Health Care System, MD, Baltimore, USA.,Department of Pathology, University of Maryland School of Medicine, MD, Baltimore, USA.,Department of Physiology, University of Maryland School of Medicine, MD, Baltimore, USA
| | - Chozha Vendan Rathinam
- Institute of Human Virology, University of Maryland School of Medicine, 725 West Lombard Street, Baltimore, MD, 21201, USA. .,Center for Stem Cell and Regenerative Medicine, University of Maryland School of Medicine, MD, 21201, Baltimore, USA.
| |
Collapse
|
49
|
N-Linked Glycans Shape Skin Immune Responses during Arthritis and Myositis after Intradermal Infection with Ross River Virus. J Virol 2022; 96:e0099922. [PMID: 36000846 PMCID: PMC9472629 DOI: 10.1128/jvi.00999-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Arthritogenic alphaviruses are mosquito-borne arboviruses that include several re-emerging human pathogens, including the chikungunya (CHIKV), Ross River (RRV), Mayaro (MAYV), and o'nyong-nyong (ONNV) virus. Arboviruses are transmitted via a mosquito bite to the skin. Herein, we describe intradermal RRV infection in a mouse model that replicates the arthritis and myositis seen in humans with Ross River virus disease (RRVD). We show that skin infection with RRV results in the recruitment of inflammatory monocytes and neutrophils, which together with dendritic cells migrate to draining lymph nodes (LN) of the skin. Neutrophils and monocytes are productively infected and traffic virus from the skin to LN. We show that viral envelope N-linked glycosylation is a key determinant of skin immune responses and disease severity. RRV grown in mammalian cells elicited robust early antiviral responses in the skin, while RRV grown in mosquito cells stimulated poorer early antiviral responses. We used glycan mass spectrometry to characterize the glycan profile of mosquito and mammalian cell-derived RRV, showing deglycosylation of the RRV E2 glycoprotein is associated with curtailed skin immune responses and reduced disease following intradermal infection. Altogether, our findings demonstrate skin infection with an arthritogenic alphavirus leads to musculoskeletal disease and envelope glycoprotein glycosylation shapes disease outcome. IMPORTANCE Arthritogenic alphaviruses are transmitted via mosquito bites through the skin, potentially causing debilitating diseases. Our understanding of how viral infection starts in the skin and how virus systemically disseminates to cause disease remains limited. Intradermal arbovirus infection described herein results in musculoskeletal pathology, which is dependent on viral envelope N-linked glycosylation. As such, intradermal infection route provides new insights into how arboviruses cause disease and could be extended to future investigations of skin immune responses following infection with other re-emerging arboviruses.
Collapse
|
50
|
Murine precursors to type 1 conventional dendritic cells induce tumor cytotoxicity and exhibit activated PD-1/PD-L1 pathway. PLoS One 2022; 17:e0273075. [PMID: 35980974 PMCID: PMC9387840 DOI: 10.1371/journal.pone.0273075] [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: 08/17/2021] [Accepted: 08/02/2022] [Indexed: 11/27/2022] Open
Abstract
The immediate precursor to murine type 1 conventional DCs (cDC1s) has recently been established and named “pre-cDC1s”. Mature CD8α+ cDC1s are recognized for suppressing graft-versus-host disease (GvHD) while promoting graft-versus-leukemia (GvL), however pre-cDC1s have not previously been investigated in the context of alloreactivity or anti-tumor responses. Characterization of pre-cDC1s, compared to CD8α+ cDC1s, found that a lower percentage of pre-cDC1s express PD-L1, yet express greater PD-L1 by MFI and a greater percent PIR-B, a GvHD-suppressing molecule. Functional assays were performed ex vivo following in vivo depletion of CD8α+ DCs to examine whether pre-cDC1s play a redundant role in alloreactivity. Proliferation assays revealed less allogeneic T-cell proliferation in the absence of CD8α+ cDC1s, with slightly greater CD8+ T-cell proliferation. Further, in the absence of CD8α+ cDC1s, stimulated CD8+ T-cells exhibited significantly less PD-1 expression compared to CD4+ T-cells, and alloreactive T-cell death was significantly lower, driven by reduced CD4+ T-cell death. Tumor-killing assays revealed that T-cells primed with CD8α-depleted DCs ex vivo induce greater killing of A20 B-cell leukemia cells, particularly when antigen (Ag) is limited. Bulk RNA sequencing revealed distinct transcriptional programs of these DCs, with pre-cDC1s exhibiting activated PD-1/PD-L1 signaling compared to CD8α+ cDC1s. These results indicate distinct T-cell-priming capabilities of murine pre-cDC1s compared to CD8α+ cDC1s ex vivo, with potentially clinically relevant implications in suppressing GvHD while promoting GvL responses, highlighting the need for greater investigation of murine pre-cDC1s.
Collapse
|