1
|
Papadas A, Lagal DJ, Dou Y, Hong D, Gibbons A, Cicala A, Huang Y, Zomalan B, Molina E, Asimakopoulos F. Protocol to identify and isolate rare murine tumor-resident dendritic cell populations for low-input transcriptomic profiling. STAR Protoc 2024; 5:103195. [PMID: 39096495 PMCID: PMC11345558 DOI: 10.1016/j.xpro.2024.103195] [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: 03/22/2024] [Revised: 05/16/2024] [Accepted: 06/22/2024] [Indexed: 08/05/2024] Open
Abstract
Conventional type 1 dendritic cells (cDC1s) are critical for innate sensing of cancer, yet they are scarce in the tumor microenvironment (TME). Here, we present a protocol to identify and isolate cDC1 subsets from murine implantable tumors for subsequent transcriptomic profiling using a flow sorting-based strategy. We describe steps for cell culture of mouse tumors, tumoral growth, dissociation and isolation of tumoral cells, extracellular staining, and cell sorting. We then detail procedures for RNA isolation, mRNA library preparation, and sequencing. For complete details on the use and execution of this protocol, please refer to Papadas et al.1.
Collapse
Affiliation(s)
- Athanasios Papadas
- Division of Blood and Marrow Transplantation, Department of Medicine, University of California, San Diego (UCSD), La Jolla, CA, USA; Moores Cancer Center, University of California, San Diego (UCSD), La Jolla, CA, USA.
| | - Daniel J Lagal
- Division of Blood and Marrow Transplantation, Department of Medicine, University of California, San Diego (UCSD), La Jolla, CA, USA; Moores Cancer Center, University of California, San Diego (UCSD), La Jolla, CA, USA
| | - Yaling Dou
- Division of Blood and Marrow Transplantation, Department of Medicine, University of California, San Diego (UCSD), La Jolla, CA, USA; Moores Cancer Center, University of California, San Diego (UCSD), La Jolla, CA, USA
| | - Duncan Hong
- Division of Blood and Marrow Transplantation, Department of Medicine, University of California, San Diego (UCSD), La Jolla, CA, USA; Moores Cancer Center, University of California, San Diego (UCSD), La Jolla, CA, USA
| | - Alicia Gibbons
- Division of Blood and Marrow Transplantation, Department of Medicine, University of California, San Diego (UCSD), La Jolla, CA, USA; Moores Cancer Center, University of California, San Diego (UCSD), La Jolla, CA, USA
| | - Alexander Cicala
- Division of Blood and Marrow Transplantation, Department of Medicine, University of California, San Diego (UCSD), La Jolla, CA, USA; Moores Cancer Center, University of California, San Diego (UCSD), La Jolla, CA, USA
| | - Yun Huang
- Division of Blood and Marrow Transplantation, Department of Medicine, University of California, San Diego (UCSD), La Jolla, CA, USA; Moores Cancer Center, University of California, San Diego (UCSD), La Jolla, CA, USA
| | - Brolyn Zomalan
- Division of Blood and Marrow Transplantation, Department of Medicine, University of California, San Diego (UCSD), La Jolla, CA, USA; Moores Cancer Center, University of California, San Diego (UCSD), La Jolla, CA, USA
| | - Elsa Molina
- Next Generation Sequencing Core, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Fotis Asimakopoulos
- Division of Blood and Marrow Transplantation, Department of Medicine, University of California, San Diego (UCSD), La Jolla, CA, USA; Moores Cancer Center, University of California, San Diego (UCSD), La Jolla, CA, USA.
| |
Collapse
|
2
|
Ullah Khan F, Khongorzul P, Gris D, Amrani A. Stat5b/Ezh2 axis governs high PD-L1 expressing tolerogenic dendritic cell subset in autoimmune diabetes. Int Immunopharmacol 2024; 133:112166. [PMID: 38678673 DOI: 10.1016/j.intimp.2024.112166] [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: 02/20/2024] [Revised: 04/18/2024] [Accepted: 04/24/2024] [Indexed: 05/01/2024]
Abstract
Dendritic cells (DCs) are specialized antigen-presenting cells that play an important role in inducing and maintaining immune tolerance. The altered distribution and/or function of DCs contributes to defective tolerance in autoimmune diseases such as type 1 diabetes (T1D). In human T1D and in NOD mouse models, DCs share some defects and are often described as less tolerogenic and excessively immunogenic. In the NOD mouse model, the autoimmune response is associated with a defect in the Stat5b signaling pathway. We have reported that expressing a constitutively active form of Stat5b in DCs of transgenic NOD mice (NOD.Stat5b-CA), re-established their tolerogenic function, restored autoimmune tolerance and conferred protection from diabetes. However, the role and molecular mechanisms of Stat5b signaling in regulating splenic conventional DCs tolerogenic signature remained unclear. In this study, we reported that, compared to immunogenic splenic DCs of NOD, splenic DCs of NOD.Stat5b-CA mice exhibited a tolerogenic profile marked by elevated PD-L1 and PD-L2 expression, reduced pro-inflammatory cytokine production, increased frequency of the cDC2 subset and decreased frequency of the cDC1 subset. This tolerogenic profile was associated with increased Ezh2 and IRF4 but decreased IRF8 expression. We also found an upregulation of PD-L1 in the cDC1 subset and high PD-L1 and PD-L2 expression in cDC2 of NOD.Stat5b-CA mice. Mechanistically, we demonstrated that Ezh2 plays an important role in the maintenance of high PD-L1 expression in cDC1 and cDC2 subsets and that Ezh2 inhibition resulted in PD-L1 but not PD-L2 downregulation which was more drastic in the cDC2 subset. Additionally, Ezh2 inhibition severely reduced the cDC2 subset and increased the cDC1 subset and Stat5b-CA.DC pro-inflammatory cytokine production. Together our data suggest that the Stat5b-Ezh2 axis is critical for the maintenance of tolerogenic high PD-L1-expressing cDC2 and autoimmune tolerance in NOD.Stat5b-CA mice.
Collapse
Affiliation(s)
- Farhan Ullah Khan
- Department of Pediatrics, Immunology Division, Faculty of Medicine and Health Sciences, Centre de Recherche du CHUS, 3001, 12th Avenue North, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Puregmaa Khongorzul
- Department of Pediatrics, Immunology Division, Faculty of Medicine and Health Sciences, Centre de Recherche du CHUS, 3001, 12th Avenue North, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Denis Gris
- Department of Pediatrics, Immunology Division, Faculty of Medicine and Health Sciences, Centre de Recherche du CHUS, 3001, 12th Avenue North, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Abdelaziz Amrani
- Department of Pediatrics, Immunology Division, Faculty of Medicine and Health Sciences, Centre de Recherche du CHUS, 3001, 12th Avenue North, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada.
| |
Collapse
|
3
|
Swarnkar G, Semenkovich NP, Arra M, Mims DK, Naqvi SK, Peterson T, Mbalaviele G, Wu CL, Abu-Amer Y. DNA hypomethylation ameliorates erosive inflammatory arthritis by modulating interferon regulatory factor-8. Proc Natl Acad Sci U S A 2024; 121:e2310264121. [PMID: 38319963 PMCID: PMC10873594 DOI: 10.1073/pnas.2310264121] [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: 06/22/2023] [Accepted: 01/08/2024] [Indexed: 02/08/2024] Open
Abstract
Epigenetic regulation plays a crucial role in the pathogenesis of autoimmune diseases such as inflammatory arthritis. DNA hypomethylating agents, such as decitabine (DAC), have been shown to dampen inflammation and restore immune homeostasis. In the present study, we demonstrate that DAC elicits potent anti-inflammatory effects and attenuates disease symptoms in several animal models of arthritis. Transcriptomic and epigenomic profiling show that DAC-mediated hypomethylation regulates a wide range of cell types in arthritis, altering the differentiation trajectories of anti-inflammatory macrophage populations, regulatory T cells, and tissue-protective synovial fibroblasts (SFs). Mechanistically, DAC-mediated demethylation of intragenic 5'-Cytosine phosphate Guanine-3' (CpG) islands of the transcription factor Irf8 (interferon regulatory factor 8) induced its re-expression and promoted its repressor activity. As a result, DAC restored joint homeostasis by resetting the transcriptomic signature of negative regulators of inflammation in synovial macrophages (MerTK, Trem2, and Cx3cr1), TREGs (Foxp3), and SFs (Pdpn and Fapα). In conclusion, we found that Irf8 is necessary for the inhibitory effect of DAC in murine arthritis and that direct expression of Irf8 is sufficient to significantly mitigate arthritis.
Collapse
Affiliation(s)
- Gaurav Swarnkar
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO63110
| | | | - Manoj Arra
- Department of Emergency Medicine, Washington University School of Medicine, St. Louis, MO63110
| | - Dorothy K. Mims
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO63110
| | - Syeda Kanwal Naqvi
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO63110
| | - Timothy Peterson
- Department of Medicine, Washington University School of Medicine, St. Louis, MO63110
- HealthSpan Technologies, Inc, St. Louis, MO63110
| | - Gabriel Mbalaviele
- Department of Medicine, Washington University School of Medicine, St. Louis, MO63110
| | - Chia-Lung Wu
- Department of Orthopedics and Physical Performance, University of Rochester, Rochester, NY14642
| | - Yousef Abu-Amer
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO63110
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO63110
- Shriners Hospital for Children, St. Louis, MO63110
| |
Collapse
|
4
|
Janssens S, Rennen S, Agostinis P. Decoding immunogenic cell death from a dendritic cell perspective. Immunol Rev 2024; 321:350-370. [PMID: 38093416 DOI: 10.1111/imr.13301] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2024]
Abstract
Dendritic cells (DCs) are myeloid cells bridging the innate and adaptive immune system. By cross-presenting tumor-associated antigens (TAAs) liberated upon spontaneous or therapy-induced tumor cell death to T cells, DCs occupy a pivotal position in the cancer immunity cycle. Over the last decades, the mechanisms linking cancer cell death to DC maturation, have been the focus of intense research. Growing evidence supports the concept that the mere transfer of TAAs during the process of cell death is insufficient to drive immunogenic DC maturation unless this process is coupled with the release of immunomodulatory signals by dying cancer cells. Malignant cells succumbing to a regulated cell death variant called immunogenic cell death (ICD), foster a proficient interface with DCs, enabling their immunogenic maturation and engagement of adaptive immunity against cancer. This property relies on the ability of ICD to exhibit pathogen-mimicry hallmarks and orchestrate the emission of a spectrum of constitutively present or de novo-induced danger signals, collectively known as damage-associated molecular patterns (DAMPs). In this review, we discuss how DCs perceive and decode danger signals emanating from malignant cells undergoing ICD and provide an outlook of the major signaling and functional consequences of this interaction for DCs and antitumor immunity.
Collapse
Affiliation(s)
- Sophie Janssens
- Laboratory for ER Stress and Inflammation, Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Sofie Rennen
- Laboratory for ER Stress and Inflammation, Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Patrizia Agostinis
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
5
|
Ly6D +Siglec-H + precursors contribute to conventional dendritic cells via a Zbtb46 +Ly6D + intermediary stage. Nat Commun 2022; 13:3456. [PMID: 35705536 PMCID: PMC9200809 DOI: 10.1038/s41467-022-31054-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/24/2022] [Indexed: 11/09/2022] Open
Abstract
Plasmacytoid and conventional dendritic cells (pDC and cDC) are generated from progenitor cells in the bone marrow and commitment to pDCs or cDC subtypes may occur in earlier and later progenitor stages. Cells within the CD11c+MHCII-/loSiglec-H+CCR9lo DC precursor fraction of the mouse bone marrow generate both pDCs and cDCs. Here we investigate the heterogeneity and commitment of subsets in this compartment by single-cell transcriptomics and high-dimensional flow cytometry combined with cell fate analysis: Within the CD11c+MHCII-/loSiglec-H+CCR9lo DC precursor pool cells expressing high levels of Ly6D and lacking expression of transcription factor Zbtb46 contain CCR9loB220hi immediate pDC precursors and CCR9loB220lo (lo-lo) cells which still generate pDCs and cDCs in vitro and in vivo under steady state conditions. cDC-primed cells within the Ly6DhiZbtb46- lo-lo precursors rapidly upregulate Zbtb46 and pass through a Zbtb46+Ly6D+ intermediate stage before acquiring cDC phenotype after cell division. Type I IFN stimulation limits cDC and promotes pDC output from this precursor fraction by arresting cDC-primed cells in the Zbtb46+Ly6D+ stage preventing their expansion and differentiation into cDCs. Modulation of pDC versus cDC output from precursors by external factors may allow for adaptation of DC subset composition at later differentiation stages.
Collapse
|
6
|
Larsen SE, Williams BD, Rais M, Coler RN, Baldwin SL. It Takes a Village: The Multifaceted Immune Response to Mycobacterium tuberculosis Infection and Vaccine-Induced Immunity. Front Immunol 2022; 13:840225. [PMID: 35359957 PMCID: PMC8960931 DOI: 10.3389/fimmu.2022.840225] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/08/2022] [Indexed: 11/18/2022] Open
Abstract
Despite co-evolving with humans for centuries and being intensely studied for decades, the immune correlates of protection against Mycobacterium tuberculosis (Mtb) have yet to be fully defined. This lapse in understanding is a major lag in the pipeline for evaluating and advancing efficacious vaccine candidates. While CD4+ T helper 1 (TH1) pro-inflammatory responses have a significant role in controlling Mtb infection, the historically narrow focus on this cell population may have eclipsed the characterization of other requisite arms of the immune system. Over the last decade, the tuberculosis (TB) research community has intentionally and intensely increased the breadth of investigation of other immune players. Here, we review mechanistic preclinical studies as well as clinical anecdotes that suggest the degree to which different cell types, such as NK cells, CD8+ T cells, γ δ T cells, and B cells, influence infection or disease prevention. Additionally, we categorically outline the observed role each major cell type plays in vaccine-induced immunity, including Mycobacterium bovis bacillus Calmette-Guérin (BCG). Novel vaccine candidates advancing through either the preclinical or clinical pipeline leverage different platforms (e.g., protein + adjuvant, vector-based, nucleic acid-based) to purposefully elicit complex immune responses, and we review those design rationales and results to date. The better we as a community understand the essential composition, magnitude, timing, and trafficking of immune responses against Mtb, the closer we are to reducing the severe disease burden and toll on human health inflicted by TB globally.
Collapse
Affiliation(s)
- Sasha E. Larsen
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States
| | - Brittany D. Williams
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States,Department of Global Health, University of Washington, Seattle, WA, United States
| | - Maham Rais
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States
| | - Rhea N. Coler
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States,Department of Global Health, University of Washington, Seattle, WA, United States,Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, United States
| | - Susan L. Baldwin
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, United States,*Correspondence: Susan L. Baldwin,
| |
Collapse
|
7
|
Early life exposure to house dust mite allergen prevents experimental allergic asthma requiring mitochondrial H 2O 2. Mucosal Immunol 2022; 15:154-164. [PMID: 34580428 PMCID: PMC8738138 DOI: 10.1038/s41385-021-00458-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 09/08/2021] [Accepted: 09/13/2021] [Indexed: 02/04/2023]
Abstract
Immune tolerance to allergens in early-life decreases the risk for asthma in later life. Here we show establishment of stable airway tolerance to the allergen, house dust mite (HDM), by exposing newborn mice repeatedly to a low dose of the allergen. Lung dendritic cells (DCs) from tolerized mice induced a low Th2 response in vitro mirroring impact of tolerance in vivo. In line with our previous finding of increased mitochondrial H2O2 production from lung DCs of mice tolerized to ovalbumin, depletion of mitochondrial H2O2 in MCAT mice abrogated HDM-induced airway tolerance (Tol) with elevated Th2 effector response, airway eosinophilia, and increased airway hyperreactivity. WT-Tol mice displayed a decrease in total, cDC1 and cDC2 subsets in the lung as compared to that in naive mice. In contrast, the lungs of MCAT-Tol mice showed 3-fold higher numbers of cDCs including those of the subsets as compared to that in WT mice. Our study demonstrates an important role of mitochondrial H2O2 in constraining lung DC numbers towards establishment of early-life airway tolerance to allergens.
Collapse
|
8
|
Papadas A, Cicala A, Kraus SG, Arauz G, Tong A, Deming D, Asimakopoulos F. Versican in Tumor Progression, Tumor–Host Interactions, and Cancer Immunotherapy. BIOLOGY OF EXTRACELLULAR MATRIX 2022:93-118. [DOI: 10.1007/978-3-030-99708-3_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
9
|
Zhang S, Chopin M, Nutt SL. Type 1 conventional dendritic cells: ontogeny, function, and emerging roles in cancer immunotherapy. Trends Immunol 2021; 42:1113-1127. [PMID: 34728143 DOI: 10.1016/j.it.2021.10.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 12/12/2022]
Abstract
Dendritic cells (DCs) are key immune sentinels that orchestrate protective immune responses against pathogens or cancers. DCs have evolved into multiple phenotypically, anatomically, and functionally distinct cell types. One of these DC types, Type 1 conventional DCs (cDC1s), are uniquely equipped to promote cytotoxic CD8+ T cell differentiation and, therefore, represent a promising target for harnessing antitumor immunity. Indeed, recent studies have highlighted the importance of cDC1s in tumor immunotherapy using immune checkpoint inhibitors. Here, we review the progress in defining the key developmental and functional attributes of cDC1s and the approaches to optimizing the potency of cDC1s for anticancer immunity.
Collapse
Affiliation(s)
- Shengbo Zhang
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Michaël Chopin
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia.
| | - Stephen L Nutt
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
10
|
Tewari A, Prabagar MG, Gibbings SL, Rawat K, Jakubzick CV. LN Monocytes Limit DC-Poly I:C Induced Cytotoxic T Cell Response via IL-10 and Induction of Suppressor CD4 T Cells. Front Immunol 2021; 12:763379. [PMID: 34691085 PMCID: PMC8527167 DOI: 10.3389/fimmu.2021.763379] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 09/20/2021] [Indexed: 12/24/2022] Open
Abstract
Every immune response has accelerators and brakes. Depending on the pathogen or injury, monocytes can play either role, promoting or resolving immunity. Poly I:C, a potent TLR3 ligand, licenses cross-presenting dendritic cells (DC1) to accelerate a robust cytotoxic T cells response against a foreign antigen. Poly I:C thus has promise as an adjuvant in cancer immunotherapy and viral subunit vaccines. Like DC1s, monocytes are also abundant in the LNs. They may act as either immune accelerators or brakes, depending on the inflammatory mediator they encounter. However, little is known about their contribution to adaptive immunity in the context of antigen and Poly I:C. Using monocyte-deficient and chimeric mice, we demonstrate that LN monocytes indirectly dampen a Poly I:C induced antigen-specific cytotoxic T cell response, exerting a “braking” function. This effect is mediated by IL-10 production and induction of suppressor CD4+ T cells. In a metastatic melanoma model, we show that a triple-combination prophylactic treatment consisting of anti-IL-10, tumor peptides and Poly I:C works because removing IL-10 counteracts the monocytic brake, resulting in significantly fewer tumors compared to mice treated with tumor peptides and Poly I:C alone. Finally, in human LN tissue, we observed that monocytes (unlike DCs) express high levels of IL-10, suggesting that anti-IL-10 may be an important addition to treatments. Overall, our data demonstrates that LN monocytes regulate the induction of a robust DC1-mediated immune response. Neutralization of either IL-10 or monocytes can augment Poly I:C-based treatments and enhance T cell cytotoxicity.
Collapse
Affiliation(s)
- Anita Tewari
- Department of Microbiology and Immunology, Geisel School of Medicine, Hanover, NH, United States
| | - Miglena G Prabagar
- Department of Pediatrics, National Jewish Health, Denver, CO, United States
| | - Sophie L Gibbings
- Department of Pediatrics, National Jewish Health, Denver, CO, United States
| | - Kavita Rawat
- Department of Microbiology and Immunology, Geisel School of Medicine, Hanover, NH, United States
| | - Claudia V Jakubzick
- Department of Microbiology and Immunology, Geisel School of Medicine, Hanover, NH, United States
| |
Collapse
|
11
|
Peng WX, Koirala P, Zhou H, Jiang J, Zhang Z, Yang L, Mo YY. Lnc-DC promotes estrogen independent growth and tamoxifen resistance in breast cancer. Cell Death Dis 2021; 12:1000. [PMID: 34697301 PMCID: PMC8546148 DOI: 10.1038/s41419-021-04288-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 09/15/2021] [Accepted: 10/07/2021] [Indexed: 12/24/2022]
Abstract
Selective estrogen receptor modulators (SERMs) such as tamoxifen have proven to be effective in the treatment of estrogen receptor (ER) positive breast cancer. However, a major obstacle for such endocrine therapy is estrogen independent growth, leading to resistance, and the underlying mechanism is not fully understood. The purpose of this study was to determine whether long non-coding RNAs (lncRNAs) are involved in regulation of estrogen independent growth and tamoxifen resistance in ER positive breast cancer. Using a CRISPR/Cas9-based SAM (synergistic activation mediator) library against a focus group of lncRNAs, we identify Lnc-DC as a candidate lncRNA. Further analysis suggests that Lnc-DC is able to reduce tamoxifen-induced apoptosis by upregulation of anti-apoptotic genes such as Bcl2 and Bcl-xL. Furthermore, Lnc-DC activates STAT3 by phosphorylation (pSTAT3Y705), and the activated STAT3 subsequently induces expression of cytokines which in turn activate STAT3, forming an autocrine loop. Clinically, upregulation of Lnc-DC is associated with poor prognosis. In particular, analysis of a tamoxifen-treated patient cohort indicates that Lnc-DC expression can predict the response to tamoxifen. Together, this study demonstrates a previously uncharacterized function of Lnc-DC/STAT3/cytokine axis in estrogen independent growth and tamoxifen resistance, and Lnc-DC may serve as a potential predictor for tamoxifen response.
Collapse
Affiliation(s)
- Wan-Xin Peng
- Center of Oncology, Department of Medical Oncology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, PR China
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA
| | - Pratirodh Koirala
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA
- Department of Biochemistry, University of Mississippi Medical Center, Jackson, MS, USA
| | - Huaixiang Zhou
- Center of Oncology, Department of Medical Oncology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, PR China
| | - Jiahong Jiang
- Center of Oncology, Department of Medical Oncology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, PR China
| | - Ziqiang Zhang
- Department of Pulmonary Medicine, Tongji Hospital, Tongji University, Shanghai, China
| | - Liu Yang
- Center of Oncology, Department of Medical Oncology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, PR China.
| | - Yin-Yuan Mo
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA.
- Department of Pharmacology/Toxicology, University of Mississippi Medical Center, Jackson, MS, USA.
| |
Collapse
|
12
|
Izumi G, Nakano H, Nakano K, Whitehead GS, Grimm SA, Fessler MB, Karmaus PW, Cook DN. CD11b + lung dendritic cells at different stages of maturation induce Th17 or Th2 differentiation. Nat Commun 2021; 12:5029. [PMID: 34413303 PMCID: PMC8377117 DOI: 10.1038/s41467-021-25307-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 07/30/2021] [Indexed: 12/12/2022] Open
Abstract
Dendritic cells (DC) in the lung that induce Th17 differentiation remain incompletely understood, in part because conventional CD11b+ DCs (cDC2) are heterogeneous. Here, we report a population of cDCs that rapidly accumulates in lungs of mice following house dust extract inhalation. These cells are Ly-6C+, are developmentally and phenotypically similar to cDC2, and strongly promote Th17 differentiation ex vivo. Single cell RNA-sequencing (scRNA-Seq) of lung cDC2 indicates 5 distinct clusters. Pseudotime analysis of scRNA-Seq data and adoptive transfer experiments with purified cDC2 subpopulations suggest stepwise developmental progression of immature Ly-6C+Ly-6A/E+ cDC2 to mature Ly-6C-CD301b+ lung resident cDC2 lacking Ccr7 expression, which then further mature into CD200+ migratory cDC2 expressing Ccr7. Partially mature Ly-6C+Ly-6A/E-CD301b- cDC2, which express Il1b, promote Th17 differentiation. By contrast, CD200+ mature cDC2 strongly induce Th2, but not Th17, differentiation. Thus, Th17 and Th2 differentiation are promoted by lung cDC2 at distinct stages of maturation.
Collapse
Affiliation(s)
- Gentaro Izumi
- Immunity, Inflammation and Disease Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC, USA
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Hideki Nakano
- Immunity, Inflammation and Disease Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC, USA.
| | - Keiko Nakano
- Immunity, Inflammation and Disease Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC, USA
| | - Gregory S Whitehead
- Immunity, Inflammation and Disease Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC, USA
| | - Sara A Grimm
- Integrative Bioinformatics Support Group, Division of Intramural Research, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC, USA
| | - Michael B Fessler
- Immunity, Inflammation and Disease Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC, USA
| | - Peer W Karmaus
- Immunity, Inflammation and Disease Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC, USA
| | - Donald N Cook
- Immunity, Inflammation and Disease Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC, USA.
| |
Collapse
|
13
|
Charpentier J, Briand F, Lelouvier B, Servant F, Azalbert V, Puel A, Christensen JE, Waget A, Branchereau M, Garret C, Lluch J, Heymes C, Brousseau E, Burcelin R, Guzylack L, Sulpice T, Grasset E. Liraglutide targets the gut microbiota and the intestinal immune system to regulate insulin secretion. Acta Diabetol 2021; 58:881-897. [PMID: 33723651 DOI: 10.1007/s00592-020-01657-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 12/09/2020] [Indexed: 01/05/2023]
Abstract
AIMS Liraglutide controls type 2 diabetes (T2D) and inflammation. Gut microbiota regulates the immune system and causes at least in part type 2 diabetes. We here evaluated whether liraglutide regulates T2D through both gut microbiota and immunity in dysmetabolic mice. METHODS Diet-induced dysmetabolic mice were treated for 14 days with intraperitoneal injection of liraglutide (100 µg/kg) or with vehicle or Exendin 4 (10 µg/kg) as controls. Various metabolic parameters, the intestinal immune cells were characterized and the 16SrDNA gene sequenced from the gut. The causal role of gut microbiota was shown using large spectrum antibiotics and by colonization of germ-free mice with the gut microbiota from treated mice. RESULTS Besides, the expected metabolic impacts liraglutide treatment induced a specific gut microbiota specific signature when compared to vehicle or Ex4-treated mice. However, liraglutide only increased glucose-induced insulin secretion, reduced the frequency of Th1 lymphocytes, and increased that of TReg in the intestine. These effects were abolished by a concomitant antibiotic treatment. Colonization of germ-free mice with gut microbiota from liraglutide-treated diabetic mice improved glucose-induced insulin secretion and regulated the intestinal immune system differently from what observed in germ-free mice colonized with microbiota from non-treated diabetic mice. CONCLUSIONS Altogether, our result demonstrated first the influence of liraglutide on gut microbiota and the intestinal immune system which could at least in part control glucose-induced insulin secretion.
Collapse
Affiliation(s)
- Julie Charpentier
- Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France
- Unité Mixte de Recherche (UMR) 1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Université Paul Sabatier (UPS), Team 2: 'Intestinal Risk Factors Diabetes, Dyslipidemia', 31432, Toulouse Cedex 4, France
| | - Francois Briand
- PHYSIOGENEX SAS Prologue Biotech, 516 Rue Pierre et Marie Curie, 31670, Labège Innopole, France
| | - Benjamin Lelouvier
- Vaiomer, Prologue Biotech, 516 Rue Pierre et Marie Curie, 31670, Labège Innopole, France
| | - Florence Servant
- Vaiomer, Prologue Biotech, 516 Rue Pierre et Marie Curie, 31670, Labège Innopole, France
| | - Vincent Azalbert
- Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France
- Unité Mixte de Recherche (UMR) 1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Université Paul Sabatier (UPS), Team 2: 'Intestinal Risk Factors Diabetes, Dyslipidemia', 31432, Toulouse Cedex 4, France
| | - Anthony Puel
- Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France
- Unité Mixte de Recherche (UMR) 1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Université Paul Sabatier (UPS), Team 2: 'Intestinal Risk Factors Diabetes, Dyslipidemia', 31432, Toulouse Cedex 4, France
| | - Jeffrey E Christensen
- Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France
- Unité Mixte de Recherche (UMR) 1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Université Paul Sabatier (UPS), Team 2: 'Intestinal Risk Factors Diabetes, Dyslipidemia', 31432, Toulouse Cedex 4, France
| | - Aurélie Waget
- Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France
- Unité Mixte de Recherche (UMR) 1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Université Paul Sabatier (UPS), Team 2: 'Intestinal Risk Factors Diabetes, Dyslipidemia', 31432, Toulouse Cedex 4, France
| | - Maxime Branchereau
- Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France
- Unité Mixte de Recherche (UMR) 1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Université Paul Sabatier (UPS), Team 2: 'Intestinal Risk Factors Diabetes, Dyslipidemia', 31432, Toulouse Cedex 4, France
| | - Céline Garret
- Vaiomer, Prologue Biotech, 516 Rue Pierre et Marie Curie, 31670, Labège Innopole, France
| | - Jérome Lluch
- Vaiomer, Prologue Biotech, 516 Rue Pierre et Marie Curie, 31670, Labège Innopole, France
| | - Christophe Heymes
- Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France
- Unité Mixte de Recherche (UMR) 1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Université Paul Sabatier (UPS), Team 2: 'Intestinal Risk Factors Diabetes, Dyslipidemia', 31432, Toulouse Cedex 4, France
| | - Emmanuel Brousseau
- PHYSIOGENEX SAS Prologue Biotech, 516 Rue Pierre et Marie Curie, 31670, Labège Innopole, France
| | - Rémy Burcelin
- Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France.
- Unité Mixte de Recherche (UMR) 1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Université Paul Sabatier (UPS), Team 2: 'Intestinal Risk Factors Diabetes, Dyslipidemia', 31432, Toulouse Cedex 4, France.
| | - Laurence Guzylack
- Neuro-Gastroenterology and Nutrition Team, Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Thierry Sulpice
- PHYSIOGENEX SAS Prologue Biotech, 516 Rue Pierre et Marie Curie, 31670, Labège Innopole, France
| | - Estelle Grasset
- Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France
- Unité Mixte de Recherche (UMR) 1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Université Paul Sabatier (UPS), Team 2: 'Intestinal Risk Factors Diabetes, Dyslipidemia', 31432, Toulouse Cedex 4, France
| |
Collapse
|
14
|
Zakharov PN, Hu H, Wan X, Unanue ER. Single-cell RNA sequencing of murine islets shows high cellular complexity at all stages of autoimmune diabetes. J Exp Med 2021; 217:151619. [PMID: 32251514 PMCID: PMC7971127 DOI: 10.1084/jem.20192362] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/11/2020] [Accepted: 03/02/2020] [Indexed: 12/22/2022] Open
Abstract
Tissue-specific autoimmune diseases are driven by activation of diverse immune cells in the target organs. However, the molecular signatures of immune cell populations over time in an autoimmune process remain poorly defined. Using single-cell RNA sequencing, we performed an unbiased examination of diverse islet-infiltrating cells during autoimmune diabetes in the nonobese diabetic mouse. The data revealed a landscape of transcriptional heterogeneity across the lymphoid and myeloid compartments. Memory CD4 and cytotoxic CD8 T cells appeared early in islets, accompanied by regulatory cells with distinct phenotypes. Surprisingly, we observed a dramatic remodeling in the islet microenvironment, in which the resident macrophages underwent a stepwise activation program. This process resulted in polarization of the macrophage subpopulations into a terminal proinflammatory state. This study provides a single-cell atlas defining the staging of autoimmune diabetes and reveals that diabetic autoimmunity is driven by transcriptionally distinct cell populations specialized in divergent biological functions.
Collapse
Affiliation(s)
- Pavel N Zakharov
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Hao Hu
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Xiaoxiao Wan
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Emil R Unanue
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
15
|
Bajana S, Thomas K, Georgescu C, Zhao Y, Wren JD, Kovats S, Sun XH. Augmenting E Protein Activity Impairs cDC2 Differentiation at the Pre-cDC Stage. Front Immunol 2020; 11:577718. [PMID: 33391258 PMCID: PMC7775562 DOI: 10.3389/fimmu.2020.577718] [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: 06/29/2020] [Accepted: 11/12/2020] [Indexed: 11/24/2022] Open
Abstract
Dendritic cell (DC) specification and differentiation are controlled by a circuit of transcription factors, which regulate the expression of DC effector genes as well as the transcription factors themselves. E proteins are a widely expressed basic helix-loop-helix family of transcription factors whose activity is suppressed by their inhibitors, ID proteins. Loss-of-function studies have demonstrated the essential role of both E and ID proteins in different aspects of DC development. In this study, we employed a gain-of-function approach to illustrate the importance of the temporal control of E protein function in maintaining balanced differentiation of conventional DC (cDC) subsets, cDC1 and cDC2. We expressed an E protein mutant, ET2, which dimerizes with endogenous E proteins to overcome inhibition by ID proteins and activate the transcription of E protein targets. Induction of ET2 expression at the hematopoietic progenitor stage led to a dramatic reduction in cDC2 precursors (pre-cDC2s) with little impact on pre-cDC1s. Consequently, we observed decreased numbers of cDC2s in the spleen and lung, as well as in FLT3L-driven bone marrow-derived DC cultures. Furthermore, in mice bearing ET2, we detected increased expression of the IRF8 transcription factor in cDC2s, in which IRF8 is normally down-regulated and IRF4 up-regulated. This aberrant expression of IRF8 induced by ET2 may contribute to the impairment of cDC2 differentiation. In addition, analyses of the transcriptomes of splenic cDC1s and cDC2s revealed that ET2 expression led to a shift, at least in part, of the transcriptional profile characteristic of cDC2s to that of cDC1. Together, these results suggest that a precise control of E protein activity is crucial for balanced DC differentiation.
Collapse
Affiliation(s)
- Sandra Bajana
- Program in Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Kevin Thomas
- Program in Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Constantin Georgescu
- Program in Genes and Human Diseases, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Ying Zhao
- Program in Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Jonathan D. Wren
- Program in Genes and Human Diseases, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Susan Kovats
- Program in Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Xiao-Hong Sun
- Program in Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| |
Collapse
|
16
|
Papadas A, Arauz G, Cicala A, Wiesner J, Asimakopoulos F. Versican and Versican-matrikines in Cancer Progression, Inflammation, and Immunity. J Histochem Cytochem 2020; 68:871-885. [PMID: 32623942 PMCID: PMC7711242 DOI: 10.1369/0022155420937098] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/04/2020] [Indexed: 12/16/2022] Open
Abstract
Versican is an extracellular matrix proteoglycan with key roles in multiple facets of cancer development, ranging from proliferative signaling, evasion of growth-suppressor pathways, regulation of cell death, promotion of neoangiogenesis, and tissue invasion and metastasis. Multiple lines of evidence implicate versican and its bioactive proteolytic fragments (matrikines) in the regulation of cancer inflammation and antitumor immune responses. The understanding of the dynamics of versican deposition/accumulation and its proteolytic turnover holds potential for the development of novel immune biomarkers as well as approaches to reset the immune thermostat of tumors, thus promoting efficacy of modern immunotherapies. This article summarizes work from several laboratories, including ours, on the role of this central matrix proteoglycan in tumor progression as well as tumor-immune cell cross-talk.
Collapse
Affiliation(s)
- Athanasios Papadas
- Division of Blood and Marrow Transplantation, Department of Medicine, Moores Cancer Center, University of California, San Diego, La Jolla, CA
- Cellular & Molecular Pathology Graduate Program, University of Wisconsin-Madison, Madison, WI
| | - Garrett Arauz
- Division of Blood and Marrow Transplantation, Department of Medicine, Moores Cancer Center, University of California, San Diego, La Jolla, CA
| | - Alexander Cicala
- Division of Blood and Marrow Transplantation, Department of Medicine, Moores Cancer Center, University of California, San Diego, La Jolla, CA
| | - Joshua Wiesner
- Division of Blood and Marrow Transplantation, Department of Medicine, Moores Cancer Center, University of California, San Diego, La Jolla, CA
| | - Fotis Asimakopoulos
- Division of Blood and Marrow Transplantation, Department of Medicine, Moores Cancer Center, University of California, San Diego, La Jolla, CA
| |
Collapse
|
17
|
Leach SM, Gibbings SL, Tewari AD, Atif SM, Vestal B, Danhorn T, Janssen WJ, Wager TD, Jakubzick CV. Human and Mouse Transcriptome Profiling Identifies Cross-Species Homology in Pulmonary and Lymph Node Mononuclear Phagocytes. Cell Rep 2020; 33:108337. [PMID: 33147458 PMCID: PMC7673261 DOI: 10.1016/j.celrep.2020.108337] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 08/15/2020] [Accepted: 10/08/2020] [Indexed: 12/24/2022] Open
Abstract
The mononuclear phagocyte (MP) system consists of macrophages, monocytes, and dendritic cells (DCs). MP subtypes play distinct functional roles in steady-state and inflammatory conditions. Although murine MPs are well characterized, their pulmonary and lymph node (LN) human homologs remain poorly understood. To address this gap, we have created a gene expression compendium across 24 distinct human and murine lung and LN MPs, along with human blood and murine spleen MPs, to serve as validation datasets. In-depth RNA sequencing identifies corresponding human-mouse MP subtypes and determines marker genes shared and divergent across species. Unexpectedly, only 13%-23% of the top 1,000 marker genes (i.e., genes not shared across species-specific MP subtypes) overlap in corresponding human-mouse MP counterparts. Lastly, CD88 in both species helps distinguish monocytes/macrophages from DCs. Our cross-species expression compendium serves as a resource for future translational studies to investigate beforehand whether pursuing specific MP subtypes or genes will prove fruitful.
Collapse
Affiliation(s)
- Sonia M Leach
- Center for Genes, Environment, and Health, National Jewish Health, Denver, CO 80206, USA; Department of Biomedical Research, National Jewish Health, Denver, CO 80206, USA
| | - Sophie L Gibbings
- Department of Pediatrics, National Jewish Health, Denver, CO 80206, USA
| | - Anita D Tewari
- Department of Microbiology and Immunology, Dartmouth College, Hanover, NH 03756, USA
| | - Shaikh M Atif
- Department of Medicine, Division of Asthma, Allergy, and Clinical Immunology, University of Colorado, Denver, CO 80045, USA
| | - Brian Vestal
- Center for Genes, Environment, and Health, National Jewish Health, Denver, CO 80206, USA; Department of Biomedical Research, National Jewish Health, Denver, CO 80206, USA
| | - Thomas Danhorn
- Center for Genes, Environment, and Health, National Jewish Health, Denver, CO 80206, USA
| | - William J Janssen
- Department of Medicine, National Jewish Health, Denver, CO 80206, USA; Division of Pulmonary Sciences and Critical Care, University of Colorado, Denver, CO 80045, USA
| | - Tor D Wager
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO 80309, USA
| | - Claudia V Jakubzick
- Department of Pediatrics, National Jewish Health, Denver, CO 80206, USA; Department of Microbiology and Immunology, Dartmouth College, Hanover, NH 03756, USA; Department of Immunology, University of Colorado, Denver Anschutz Campus, Denver, CO 80045, USA.
| |
Collapse
|
18
|
Wong E, Montoya B, Stotesbury C, Ferez M, Xu RH, Sigal LJ. Langerhans Cells Orchestrate the Protective Antiviral Innate Immune Response in the Lymph Node. Cell Rep 2020; 29:3047-3059.e3. [PMID: 31801072 PMCID: PMC6927544 DOI: 10.1016/j.celrep.2019.10.118] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 09/17/2019] [Accepted: 10/29/2019] [Indexed: 12/22/2022] Open
Abstract
During disseminating viral infections, a swift innate immune response (IIR) in the draining lymph node (dLN) that restricts systemic viral spread is critical for optimal resistance to disease. However, it is unclear how this IIR is orchestrated. We show that after footpad infection of mice with ectromelia virus, dendritic cells (DCs) highly expressing major histocompatibility complex class II (MHC class IIhi DCs), including CD207+ epidermal Langerhans cells (LCs), CD103+CD207+ double-positive dermal DCs (DP-DCs), and CD103−CD207− double-negative dermal DCs (DN-DCs) migrate to the dLN from the skin carrying virus. MHC class IIhi DCs, predominantly LCs and DP-DCs, are the first cells upregulating IIR cytokines in the dLN. Preventing MHC class IIhi DC migration or depletion of LCs, but not DP-DC deficiency, suppresses the IIR in the dLN and results in high viral lethality. Therefore, LCs are the architects of an early IIR in the dLN that is critical for optimal resistance to a disseminating viral infection. Wong et al. show that by producing chemokines that recruit monocytes and by upregulating NKG2D ligands that activate ILCs, Langerhans cells are responsible for the innate immune cascade in the lymph node that is critical for survival of infection with a disseminating virus.
Collapse
Affiliation(s)
- Eric Wong
- Department of Microbiology and Immunology, Thomas Jefferson University, BLSB 709 233 South 10(th) Street, Philadelphia, PA 19107, USA
| | - Brian Montoya
- Department of Microbiology and Immunology, Thomas Jefferson University, BLSB 709 233 South 10(th) Street, Philadelphia, PA 19107, USA
| | - Colby Stotesbury
- Department of Microbiology and Immunology, Thomas Jefferson University, BLSB 709 233 South 10(th) Street, Philadelphia, PA 19107, USA
| | - Maria Ferez
- Department of Microbiology and Immunology, Thomas Jefferson University, BLSB 709 233 South 10(th) Street, Philadelphia, PA 19107, USA
| | - Ren-Huan Xu
- Immune Cell Development and Host Defense Program, Research Institute of Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| | - Luis J Sigal
- Department of Microbiology and Immunology, Thomas Jefferson University, BLSB 709 233 South 10(th) Street, Philadelphia, PA 19107, USA.
| |
Collapse
|
19
|
Liu Y, Sun P, Zhao Y, Liu B. The role of long non-coding RNAs and downstream signaling pathways in leukemia progression. Hematol Oncol 2020; 39:27-40. [PMID: 32621547 DOI: 10.1002/hon.2776] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/22/2020] [Accepted: 06/25/2020] [Indexed: 01/17/2023]
Abstract
The study of long non-coding RNAs (lncRNA) is a newly established field and our knowledge about them is rapidly growing. These kinds of RNAs are unchanged parts of the genome throughout evolution, that modulate cell growth, differentiation, and apoptosis during diverse physiological and pathological processes including leukemia development. They have the capability to be useful biomarkers for the diagnosis, clinical typing, prognosis, as well as potential therapeutic targets. In this study, we summarized the role of lncRNAs in the expression and function of white blood cells and oncogenic transformation into four main types of leukemia.
Collapse
Affiliation(s)
- Yadong Liu
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Penghao Sun
- Department of Andrology, The First Hospital of Jilin University, Changchun, China
| | - Yuhao Zhao
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Bin Liu
- Department of Hand Surgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
20
|
Barut GT, Lischer HEL, Bruggmann R, Summerfield A, Talker SC. Transcriptomic profiling of bovine blood dendritic cells and monocytes following TLR stimulation. Eur J Immunol 2020; 50:1691-1711. [PMID: 32592404 DOI: 10.1002/eji.202048643] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 06/11/2020] [Accepted: 06/26/2020] [Indexed: 11/06/2022]
Abstract
Dendritic cells (DC) and monocytes are vital for the initiation of innate and adaptive immune responses. Recently, we identified bona fide DC subsets in blood of cattle, revealing subset- and species-specific transcription of toll-like receptors (TLR). In the present study, we analyzed phenotypic and transcriptional responses of bovine DC subsets and monocytes to in vitro stimulation with four to six different TLR ligands. Bovine DC subsets, especially plasmacytoid DC (pDC), showed a clear increase of CCR7, CD25, CD40, CD80, CD86, and MHC-II expression both on mRNA and protein level. Flow cytometric detection of p38 MAPK phosphorylation 15 min after stimulation confirmed activation of DC subsets and monocytes in accordance with TLR gene expression. Whole-transcriptome sequencing of sorted and TLR-stimulated subsets revealed potential ligand- and subset-specific regulation of genes associated with inflammation, T-cell co-stimulation, migration, metabolic reprogramming, and antiviral activity. Gardiquimod was found to evoke strong responses both in DC subsets and monocytes, while Poly(I:C) and CpG preferentially triggered responses in cDC1 and pDC, respectively. This in-depth analysis of ligand responsiveness is essential for the rational design of vaccine adjuvants in cattle, and provides a solid basis for comparative studies on DC and monocyte biology across species.
Collapse
Affiliation(s)
- G Tuba Barut
- Institute of Virology and Immunology, Bern & Mittelhäusern, Switzerland.,Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Heidi E L Lischer
- Interfaculty Bioinformatics Unit and Swiss Institute of Bioinformatics, University of Bern, Bern, Switzerland
| | - Rémy Bruggmann
- Interfaculty Bioinformatics Unit and Swiss Institute of Bioinformatics, University of Bern, Bern, Switzerland
| | - Artur Summerfield
- Institute of Virology and Immunology, Bern & Mittelhäusern, Switzerland.,Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Stephanie C Talker
- Institute of Virology and Immunology, Bern & Mittelhäusern, Switzerland.,Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
21
|
Targeting Dendritic Cells with Antigen-Delivering Antibodies for Amelioration of Autoimmunity in Animal Models of Multiple Sclerosis and Other Autoimmune Diseases. Antibodies (Basel) 2020; 9:antib9020023. [PMID: 32549343 PMCID: PMC7345927 DOI: 10.3390/antib9020023] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/22/2020] [Accepted: 04/30/2020] [Indexed: 02/07/2023] Open
Abstract
The specific targeting of dendritic cells (DCs) using antigen-delivering antibodies has been established to be a highly efficient protocol for the induction of tolerance and protection from autoimmune processes in experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis (MS), as well as in some other animal disease models. As the specific mechanisms of such induced tolerance are being investigated, the newly gained insights may also possibly help to design effective treatments for patients. Here we review approaches applied for the amelioration of autoimmunity in animal models based on antibody-mediated targeting of self-antigens to DCs. Further, we discuss relevant mechanisms of immunological tolerance that underlie such approaches, and we also offer some future perspectives for the application of similar methods in certain related disease settings such as transplantation.
Collapse
|
22
|
Lecoeur H, Rosazza T, Kokou K, Varet H, Coppée JY, Lari A, Commère PH, Weil R, Meng G, Milon G, Späth GF, Prina E. Leishmania amazonensis Subverts the Transcription Factor Landscape in Dendritic Cells to Avoid Inflammasome Activation and Stall Maturation. Front Immunol 2020; 11:1098. [PMID: 32582184 PMCID: PMC7295916 DOI: 10.3389/fimmu.2020.01098] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 05/06/2020] [Indexed: 12/18/2022] Open
Abstract
Leishmania parasites are the causative agents of human leishmaniases. They infect professional phagocytes of their mammalian hosts, including dendritic cells (DCs) that are essential for the initiation of adaptive immune responses. These immune functions strictly depend on the DC's capacity to differentiate from immature, antigen-capturing cells to mature, antigen-presenting cells—a process accompanied by profound changes in cellular phenotype and expression profile. Only little is known on how intracellular Leishmania affects this important process and DC transcriptional regulation. Here, we investigate these important open questions analyzing phenotypic, cytokine profile and transcriptomic changes in murine, immature bone marrow-derived DCs (iBMDCs) infected with antibody-opsonized and non-opsonized Leishmania amazonensis (L.am) amastigotes. DCs infected by non-opsonized amastigotes remained phenotypically immature whereas those infected by opsonized parasites displayed a semi-mature phenotype. The low frequency of infected DCs in culture led us to use DsRed2-transgenic parasites allowing for the enrichment of infected BMDCs by FACS. Sorted infected DCs were then subjected to transcriptomic analyses using Affymetrix GeneChip technology. Independent of parasite opsonization, Leishmania infection induced expression of genes related to key DC processes involved in MHC Class I-restricted antigen presentation and alternative NF-κB activation. DCs infected by non-opsonized parasites maintained an immature phenotype and showed a small but significant down-regulation of gene expression related to pro-inflammatory TLR signaling, the canonical NF-kB pathway and the NLRP3 inflammasome. This transcriptomic profile was further enhanced in DCs infected with opsonized parasites that displayed a semi-mature phenotype despite absence of inflammasome activation. This paradoxical DC phenotype represents a Leishmania-specific signature, which to our knowledge has not been observed with other opsonized infectious agents. In conclusion, systems-analyses of our transcriptomics data uncovered important and previously unappreciated changes in the DC transcription factor landscape, thus revealing a novel Leishmania immune subversion strategy directly acting on transcriptional control of gene expression. Our data raise important questions on the dynamic and reciprocal interplay between trans-acting and epigenetic regulators in establishing permissive conditions for intracellular Leishmania infection and polarization of the immune response.
Collapse
Affiliation(s)
- Hervé Lecoeur
- Institut Pasteur, INSERM U1201, Unité de Parasitologie Moléculaire et Signalisation, Département des Parasites et Insectes Vecteurs, Paris, France.,Pasteur Institute of Shanghai, Innate Immunity Unit, Key Laboratory of Molecular Virology and Immunology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,Pasteur International Unit "Inflammation and Leishmania Infection", Paris, France
| | - Thibault Rosazza
- Institut Pasteur, INSERM U1201, Unité de Parasitologie Moléculaire et Signalisation, Département des Parasites et Insectes Vecteurs, Paris, France.,Pasteur International Unit "Inflammation and Leishmania Infection", Paris, France
| | - Kossiwa Kokou
- Institut Pasteur, INSERM U1201, Unité de Parasitologie Moléculaire et Signalisation, Département des Parasites et Insectes Vecteurs, Paris, France.,Pasteur Institute of Shanghai, Innate Immunity Unit, Key Laboratory of Molecular Virology and Immunology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,Pasteur International Unit "Inflammation and Leishmania Infection", Paris, France
| | - Hugo Varet
- Hub de Bioinformatique et Biostatistique - Département Biologie Computationnelle, Institut Pasteur, USR 3756 CNRS, Paris, France
| | - Jean-Yves Coppée
- Institut Pasteur - Transcriptome and Epigenome Platform - Biomics Pole - C2RT, Paris, France
| | - Arezou Lari
- Systems Biomedicine Unit, Institut Pasteur of Iran, Teheran, Iran
| | | | - Robert Weil
- Sorbonne Universités, Institut National de la Santé et de la Recherche Médicale (Inserm, UMR1135), Centre National de la Recherche Scientifique (CNRS, ERL8255), Centre d'Immunologie et des Maladies Infectieuses CIMI, Paris, France
| | - Guangxun Meng
- Pasteur Institute of Shanghai, Innate Immunity Unit, Key Laboratory of Molecular Virology and Immunology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,Pasteur International Unit "Inflammation and Leishmania Infection", Paris, France
| | - Genevieve Milon
- Institut Pasteur, Laboratoire Immunophysiologie et Parasitisme, Département des Parasites et Insectes Vecteurs, Paris, France
| | - Gerald F Späth
- Institut Pasteur, INSERM U1201, Unité de Parasitologie Moléculaire et Signalisation, Département des Parasites et Insectes Vecteurs, Paris, France.,Pasteur International Unit "Inflammation and Leishmania Infection", Paris, France
| | - Eric Prina
- Institut Pasteur, INSERM U1201, Unité de Parasitologie Moléculaire et Signalisation, Département des Parasites et Insectes Vecteurs, Paris, France.,Pasteur International Unit "Inflammation and Leishmania Infection", Paris, France
| |
Collapse
|
23
|
Pandey VK, Shankar BS. Radiation-induced augmentation in dendritic cell function is mediated by apoptotic bodies/STAT5/Zbtb46 signaling. Int J Radiat Biol 2020; 96:988-998. [PMID: 32396024 DOI: 10.1080/09553002.2020.1767818] [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: 10/24/2022]
Abstract
Purpose: To evaluate the effect of ionizing radiation (IR) exposure on differentiation and maturation of dendritic cells (DC).Materials and methods: Bone marrow progenitor cells irradiated in vitro or isolated from mice exposed to whole body or localized tumor irradiation were differentiated into DC. Phenotypic maturation of DC was characterized by labeling with specific antibodies and flow cytometry analysis. Cytokines were estimated by ELISA.Results: Splenic and bone marrow-derived DC (BMDC) from tumor-bearing mice exposed to localized irradiation showed abrogation of tumor-induced immunosuppression. This was not due to the effect of radiation on tumor cells as DC derived from normal mice exposed to whole-body irradiation (WBI) also showed increase in immune-activating potential of DC. This was observed in terms of increased phenotypic and functional activation of DCs. This phenomenon was also recapitulated if DC were differentiated from in vitro irradiated progenitor cells and was found to be due to STAT5/Zbtb46 signaling mediated by the irradiation-induced apoptotic bodies (ABs). When these ABs were depleted using annexin-beads, these effects were reversed confirming the involvement of this pathway. The role of ABs was further validated in DC derived from mice exposed to WBI in adaptive response experiments with 0.1 Gy priming dose prior to 2 Gy challenge dose. A corresponding reduction in DC maturation markers was observed with decrease in apoptosis in vivo. Further, these DCs derived from irradiated progenitors (IP) could resist the suppressive effects of tumor conditioned medium (TCM) and had increased immune-activating potential as seen in the tumor-bearing mice.Conclusions: Though radiation is the most commonly used therapeutic modality for cancer, its effects on dendritic cell differentiation is not completely understood. We demonstrate here for the first time that exposure to select doses of IR can increase immune-activating potential of DC through ABs. This can have implications in selection of appropriate doses of IR during radiotherapy of cancer patients.
Collapse
Affiliation(s)
- Vipul K Pandey
- Immunology Section, Radiation Biology & Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Mumbai, India
| | - Bhavani S Shankar
- Immunology Section, Radiation Biology & Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Mumbai, India.,Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
24
|
Lee J, Zhang J, Chung YJ, Kim JH, Kook CM, González-Navajas JM, Herdman DS, Nürnberg B, Insel PA, Corr M, Mo JH, Tao A, Yasuda K, Rifkin IR, Broide DH, Sciammas R, Webster NJG, Raz E. Inhibition of IRF4 in dendritic cells by PRR-independent and -dependent signals inhibit Th2 and promote Th17 responses. eLife 2020; 9:e49416. [PMID: 32014112 PMCID: PMC7000221 DOI: 10.7554/elife.49416] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 01/17/2020] [Indexed: 12/13/2022] Open
Abstract
Cyclic AMP (cAMP) is involved in many biological processes but little is known regarding its role in shaping immunity. Here we show that cAMP-PKA-CREB signaling (a pattern recognition receptor [PRR]-independent mechanism) regulates conventional type-2 Dendritic Cells (cDC2s) in mice and reprograms their Th17-inducing properties via repression of IRF4 and KLF4, transcription factors essential for cDC2-mediated Th2 induction. In mice, genetic loss of IRF4 phenocopies the effects of cAMP on Th17 induction and restoration of IRF4 prevents the cAMP effect. Moreover, curdlan, a PRR-dependent microbial product, activates CREB and represses IRF4 and KLF4, resulting in a pro-Th17 phenotype of cDC2s. These in vitro and in vivo results define a novel signaling pathway by which cDC2s display plasticity and provide a new molecular basis for the classification of novel cDC2 and cDC17 subsets. The findings also reveal that repressing IRF4 and KLF4 pathway can be harnessed for immuno-regulation.
Collapse
Affiliation(s)
- Jihyung Lee
- Department of MedicineUniversity of California San DiegoSan DiegoUnited States
| | - Junyan Zhang
- Department of MedicineUniversity of California San DiegoSan DiegoUnited States
- The Second Affiliated Hospital of Guangzhou Medical University (GMU), The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical ImmunologyGuangzhouChina
- Center for Immunology, Inflammation and Immune-mediated disease, GMUGuangzhouChina
| | - Young-Jun Chung
- Department of MedicineUniversity of California San DiegoSan DiegoUnited States
- Department of Otorhinolaryngology-Head and Neck SurgeryDankook University College of MedicineChungnamRepublic of Korea
| | - Jun Hwan Kim
- Department of MedicineUniversity of California San DiegoSan DiegoUnited States
| | - Chae Min Kook
- Department of MedicineUniversity of California San DiegoSan DiegoUnited States
| | - José M González-Navajas
- Center for Immunology, Inflammation and Immune-mediated disease, GMUGuangzhouChina
- Alicante Institute for Health and Biomedical Research (ISABIAL - FISABIO)AlicanteSpain
- Networked Biomedical Research Center for Hepatic and Digestive Diseases (CIBERehd)Institute of Health Carlos IIIMadridSpain
| | - David S Herdman
- Department of MedicineUniversity of California San DiegoSan DiegoUnited States
| | - Bernd Nürnberg
- Department of Pharmacology and Experimental TherapyUniversity of TübingenTübingenGermany
| | - Paul A Insel
- Department of MedicineUniversity of California San DiegoSan DiegoUnited States
- Department of PharmacologyUniversity of California San DiegoSan DiegoUnited States
| | - Maripat Corr
- Department of MedicineUniversity of California San DiegoSan DiegoUnited States
| | - Ji-Hun Mo
- Department of Otorhinolaryngology-Head and Neck SurgeryDankook University College of MedicineChungnamRepublic of Korea
| | - Ailin Tao
- The Second Affiliated Hospital of Guangzhou Medical University (GMU), The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical ImmunologyGuangzhouChina
- Center for Immunology, Inflammation and Immune-mediated disease, GMUGuangzhouChina
| | - Kei Yasuda
- Boston University School of MedicineBostonUnited States
| | - Ian R Rifkin
- Boston University School of MedicineBostonUnited States
- VA Boston Healthcare SystemBostonUnited States
| | - David H Broide
- Department of MedicineUniversity of California San DiegoSan DiegoUnited States
| | - Roger Sciammas
- Center for Comparative MedicineUniversity of California, DavisDavisUnited States
| | - Nicholas JG Webster
- Department of MedicineUniversity of California San DiegoSan DiegoUnited States
- VA San Diego Healthcare SystemSan DiegoUnited States
| | - Eyal Raz
- Department of MedicineUniversity of California San DiegoSan DiegoUnited States
- Center for Immunology, Inflammation and Immune-mediated disease, GMUGuangzhouChina
| |
Collapse
|
25
|
Papadas A, Asimakopoulos F. Versican in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1272:55-72. [PMID: 32845502 DOI: 10.1007/978-3-030-48457-6_4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Versican is an extracellular matrix proteoglycan with nonredundant roles in diverse biological and cellular processes, ranging from embryonic development to adult inflammation and cancer. Versican is essential for cardiovascular morphogenesis, neural crest migration, and skeletal development during embryogenesis. In the adult, versican acts as an inflammation "amplifier" and regulator of immune cell activation and cytokine production. Increased versican expression has been observed in a wide range of malignant tumors and has been associated with poor patient outcomes. The main sources of versican production in the tumor microenvironment include accessory cells (myeloid cells and stromal components) and, in some contexts, the tumor cells themselves. Versican has been implicated in several classical hallmarks of cancer such as proliferative signaling, evasion of growth suppressor signaling, resistance to cell death, angiogenesis, and tissue invasion and metastasis. More recently, versican has been implicated in escape from tumor immune surveillance, e.g., through dendritic cell dysfunction. Versican's multiple contributions to benign and malignant biological processes are further diversified through the generation of versican-derived bioactive proteolytic fragments (matrikines), with versikine being the most studied to date. Versican and versican-derived matrikines hold promise as targets in the management of inflammatory and malignant conditions as well as in the development of novel predictive and prognostic biomarkers.
Collapse
Affiliation(s)
- Athanasios Papadas
- Department of Medicine, Division of Blood and Marrow Transplantation, University of California San Diego (UCSD), Moores Cancer Center, La Jolla, CA, USA. .,University of Wisconsin-Madison, Cellular and Molecular Pathology Program, Madison, WI, USA.
| | - Fotis Asimakopoulos
- Department of Medicine, Division of Blood and Marrow Transplantation, University of California San Diego (UCSD), Moores Cancer Center, La Jolla, CA, USA
| |
Collapse
|
26
|
Jahchan NS, Mujal AM, Pollack JL, Binnewies M, Sriram V, Reyno L, Krummel MF. Tuning the Tumor Myeloid Microenvironment to Fight Cancer. Front Immunol 2019; 10:1611. [PMID: 31402908 PMCID: PMC6673698 DOI: 10.3389/fimmu.2019.01611] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 06/27/2019] [Indexed: 12/14/2022] Open
Abstract
The tumor microenvironment (TME) of diverse cancer types is often characterized by high levels of infiltrating myeloid cells including monocytes, macrophages, dendritic cells, and granulocytes. These cells perform a variety of functions in the TME, varying from immune suppressive to immune stimulatory roles. In this review, we summarize the different myeloid cell populations in the TME and the intratumoral myeloid targeting approaches that are being clinically investigated, and discuss strategies that identify new myeloid subpopulations within the TME. The TME therapies include agents that modulate the functional activities of myeloid populations, that impact recruitment and survival of myeloid subpopulations, and that functionally reprogram or activate myeloid populations. We discuss the benefits, limitations and potential side effects of these therapeutic approaches.
Collapse
Affiliation(s)
| | - Adriana M. Mujal
- Department of Pathology, University of California, San Francisco, San Francisco, CA, United States
| | | | | | | | - Leonard Reyno
- Pionyr Immunotherapeutics, South San Francisco, CA, United States
| | - Matthew F. Krummel
- Department of Pathology, University of California, San Francisco, San Francisco, CA, United States
- ImmunoX Initiative, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
27
|
Lu E, Cyster JG. G-protein coupled receptors and ligands that organize humoral immune responses. Immunol Rev 2019; 289:158-172. [PMID: 30977196 PMCID: PMC6464390 DOI: 10.1111/imr.12743] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 01/22/2019] [Indexed: 12/26/2022]
Abstract
B-cell responses are dynamic processes that depend on multiple types of interactions. Rare antigen-specific B cells must encounter antigen and specialized systems are needed-unique to each lymphoid tissue type-to ensure this happens efficiently. Lymphoid tissue barrier cells act to ensure that pathogens, while being permitted entry for B-cell recognition, are blocked from replication or dissemination. T follicular helper (Tfh) cells often need to be primed by dendritic cells before supporting B-cell responses. For most responses, antigen-specific helper T cells and B cells need to interact, first to initiate clonal expansion and the plasmablast response, and later to support the germinal center (GC) response. Newly formed plasma cells need to travel to supportive niches. GC B cells must become confined to the follicle center, organize into dark and light zones, and interact with Tfh cells. Memory B cells need to be positioned for rapid responses following reinfection. Each of these events requires the actions of multiple G-protein coupled receptors (GPCRs) and their ligands, including chemokines and lipid mediators. This review will focus on the guidance cue code underlying B-cell immunity, with an emphasis on findings from our laboratory and on newer advances in related areas. We will discuss our recent identification of geranylgeranyl-glutathione as a ligand for P2RY8. Our goal is to provide the reader with a focused knowledge about the GPCRs guiding B-cell responses and how they might be therapeutic targets, while also providing examples of how multiple types of GPCRs can cooperate or act iteratively to control cell behavior.
Collapse
Affiliation(s)
- Erick Lu
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California San Francisco, San Francisco, California
| | - Jason G Cyster
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California San Francisco, San Francisco, California
| |
Collapse
|
28
|
Yu H, Tian Y, Wang Y, Mineishi S, Zhang Y. Dendritic Cell Regulation of Graft-Vs.-Host Disease: Immunostimulation and Tolerance. Front Immunol 2019; 10:93. [PMID: 30774630 PMCID: PMC6367268 DOI: 10.3389/fimmu.2019.00093] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 01/14/2019] [Indexed: 12/12/2022] Open
Abstract
Graft-vs.-host disease (GVHD) remains a significant cause of morbidity and mortality after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Significant progresses have been made in defining the dichotomous role of dendritic cells (DCs) in the development of GVHD. Host-derived DCs are important to elicit allogeneic T cell responses, whereas certain donor-types of DCs derived from newly engrafted hematopoietic stem/progenitor cells (HSPCs) can amply this graft-vs.-host reaction. In contrast, some DCs also play non-redundant roles in mediating immune tolerance. They induce apoptotic deletion of host-reactive donor T cells while promoting expansion and function of regulatory T cells (Treg). Unfortunately, this tolerogenic effect of DCs is impaired during GVHD. Severe GVHD in patients subject to allo-HSCT is associated with significantly decreased number of circulating peripheral blood DCs during engraftment. Existing studies reveal that GVHD causes delayed reconstitution of donor DCs from engrafted HSPCs, impairs the antigen presentation function of newly generated DCs and reduces the capacity of DCs to regulate Treg. The present review will discuss the importance of DCs in alloimmunity and the mechanism underlying DC reconstitution after allo-HSCT.
Collapse
Affiliation(s)
- Hongshuang Yu
- Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA, United States
| | - Yuanyuan Tian
- Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA, United States
| | - Ying Wang
- Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA, United States
| | - Shin Mineishi
- Department of Medicine, Pennsylvania State University, Hershey, PA, United States
| | - Yi Zhang
- Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA, United States,Department of Microbiology & Immunology, Temple University, Philadelphia, PA, United States,*Correspondence: Yi Zhang
| |
Collapse
|
29
|
Mansouri S, Patel S, Katikaneni DS, Blaauboer SM, Wang W, Schattgen S, Fitzgerald K, Jin L. Immature lung TNFR2 - conventional DC 2 subpopulation activates moDCs to promote cyclic di-GMP mucosal adjuvant responses in vivo. Mucosal Immunol 2019; 12:277-289. [PMID: 30327534 PMCID: PMC6301145 DOI: 10.1038/s41385-018-0098-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 09/19/2018] [Accepted: 09/25/2018] [Indexed: 02/04/2023]
Abstract
Cyclic dinucleotides (CDNs), including cyclic di-GMP (CDG), are promising vaccine adjuvants in preclinical/clinical trials. The in vivo mechanisms of CDNs are not clear. Here we investigated the roles of lung DC subsets in promoting CDG mucosal adjuvant responses in vivo. Using genetically modified mice and adoptive cell transfer, we identified lung conventional DC 2 (cDC2) as the central player in CDG mucosal responses. We further identified two functionally distinct lung cDC2 subpopulations: TNFR2+pRelB+ and TNFR2-pRelB- cDC2. The TNFR2+ cDC2 were mature and migratory upon intranasal CDG administration while the TNFR2- cDC2 were activated but not mature. Adoptive cell transfer showed that TNFR2- cDC2 mediate the antibody responses of CDG, while the TNFR2+ cDC2 generate Th1/17 responses. Mechanistically, immature TNFR2- cDC2 activate monocyte-derived DCs (moDCs), which do not take up intranasally administered CDG. moDCs promote CDG-induced generation of T follicular helper- and germinal center B cells in the lungs. Our data revealed a previously undescribed in vivo mode of DCs action, whereby an immature lung TNFR2- cDC2 subpopulation directs the non-migratory moDCs to generate CDG mucosal responses in the lung.
Collapse
Affiliation(s)
- Samira Mansouri
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, 12208, USA
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Seema Patel
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Divya S Katikaneni
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Steven M Blaauboer
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, 12208, USA
| | - Wei Wang
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Stefan Schattgen
- Program in Innate Immunity, Division of Infectious Disease and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Katherine Fitzgerald
- Program in Innate Immunity, Division of Infectious Disease and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Lei Jin
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
30
|
Monasterio G, Guevara J, Ibarra JP, Castillo F, Díaz-Zúñiga J, Alvarez C, Cafferata EA, Vernal R. Immunostimulatory activity of low-molecular-weight hyaluronan on dendritic cells stimulated with Aggregatibacter actinomycetemcomitans or Porphyromonas gingivalis. Clin Oral Investig 2018; 23:1887-1894. [PMID: 30225677 DOI: 10.1007/s00784-018-2641-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 09/11/2018] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Periodontitis is a chronic inflammatory disease characterized by tooth-supporting tissue destruction, which is elicited by the host's immune response triggered against periodonto-pathogen bacteria. During periodontal tissue destruction, extracellular matrix components are metabolized and fragmented. Some extracellular matrix component-derived fragments, such as low-molecular-weight hyaluronan (LMW-HA), have potent immunogenic potential, playing a role as damage-associated molecular patterns (DAMPs) during activation of immune cells. Dendritic cells (DCs) play a central role in the host's immune response displayed during periodontitis; thus, this study aimed to analyze whether LMW-HA has an immunostimulatory activity on DCs when stimulated with periodonto-pathogen bacteria. MATERIALS AND METHODS LMW-HA-treated and non-treated DCs were stimulated with Aggregatibacter actinomycetemcomitans or Porphyromonas gingivalis and the mRNA expression for cytokines tumor necrosis factor-α (TNF-alpha), interleukin-1β (IL-1B), interleukin-6 (IL-6), and interleukin-23 (IL-23A) was quantified by RT-qPCR. In addition, transcription factors interferon regulatory factor 4 (IRF4), interferon regulatory factor 8 (IRF8), neurogenic locus notch homolog protein 2 (NOTCH2), and basic leucine zipper ATF-like transcription factor 3 (BATF3), involved in DC activation, were analyzed. RESULTS Higher expression levels of TNF-alpha, IL-1B, IL-6, and IL-23A were detected in LMW-HA-treated DCs after bacterial infection, as compared with non-treated DCs. When LMW-HA-treated DCs were infected with A. actinomycetemcomitans, higher levels of IRF4, NOTCH2, and BATF3 were detected compared with non-treated cells; whereas against P. gingivalis infection, increased levels of IRF4 and NOTCH2 were detected. CONCLUSION LMW-HA plays an immunostimulatory role on the immune response triggered by DCs during infection with A. actinomycetemcomitans or P. gingivalis. CLINICAL RELEVANCE Detection of extracellular matrix component-derived fragments produced during periodontal tissue destruction, such as LMW-HA, could explain at least partly unsuccessful periodontal treatment and the chronicity of the disease.
Collapse
Affiliation(s)
- Gustavo Monasterio
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Sergio Livingstone Pohlhammer 943, 8380492, Independencia, Santiago, Chile
| | - José Guevara
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Sergio Livingstone Pohlhammer 943, 8380492, Independencia, Santiago, Chile
| | - Juan Pablo Ibarra
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Sergio Livingstone Pohlhammer 943, 8380492, Independencia, Santiago, Chile
| | - Francisca Castillo
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Sergio Livingstone Pohlhammer 943, 8380492, Independencia, Santiago, Chile
| | - Jaime Díaz-Zúñiga
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Sergio Livingstone Pohlhammer 943, 8380492, Independencia, Santiago, Chile
| | - Carla Alvarez
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Sergio Livingstone Pohlhammer 943, 8380492, Independencia, Santiago, Chile
| | - Emilio A Cafferata
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Sergio Livingstone Pohlhammer 943, 8380492, Independencia, Santiago, Chile.,Faculty of Dentistry, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Rolando Vernal
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Sergio Livingstone Pohlhammer 943, 8380492, Independencia, Santiago, Chile. .,Dentistry Unit, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago, Chile.
| |
Collapse
|
31
|
Fang P, Li X, Dai J, Cole L, Camacho JA, Zhang Y, Ji Y, Wang J, Yang XF, Wang H. Immune cell subset differentiation and tissue inflammation. J Hematol Oncol 2018; 11:97. [PMID: 30064449 PMCID: PMC6069866 DOI: 10.1186/s13045-018-0637-x] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 07/02/2018] [Indexed: 02/07/2023] Open
Abstract
Immune cells were traditionally considered as major pro-inflammatory contributors. Recent advances in molecular immunology prove that immune cell lineages are composed of different subsets capable of a vast array of specialized functions. These immune cell subsets share distinct duties in regulating innate and adaptive immune functions and contribute to both immune activation and immune suppression responses in peripheral tissue. Here, we summarized current understanding of the different subsets of major immune cells, including T cells, B cells, dendritic cells, monocytes, and macrophages. We highlighted molecular characterization, frequency, and tissue distribution of these immune cell subsets in human and mice. In addition, we described specific cytokine production, molecular signaling, biological functions, and tissue population changes of these immune cell subsets in both cardiovascular diseases and cancers. Finally, we presented a working model of the differentiation of inflammatory mononuclear cells, their interaction with endothelial cells, and their contribution to tissue inflammation. In summary, this review offers an updated and comprehensive guideline for immune cell development and subset differentiation, including subset characterization, signaling, modulation, and disease associations. We propose that immune cell subset differentiation and its complex interaction within the internal biological milieu compose a “pathophysiological network,” an interactive cross-talking complex, which plays a critical role in the development of inflammatory diseases and cancers.
Collapse
Affiliation(s)
- Pu Fang
- Center for Metabolic Disease Research, Lewis Kats School of Medicine, Temple University, Medical Education and Research Building, Room 1060, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Xinyuan Li
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jin Dai
- Center for Metabolic Disease Research, Lewis Kats School of Medicine, Temple University, Medical Education and Research Building, Room 1060, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Lauren Cole
- Center for Metabolic Disease Research, Lewis Kats School of Medicine, Temple University, Medical Education and Research Building, Room 1060, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Javier Andres Camacho
- Center for Metabolic Disease Research, Lewis Kats School of Medicine, Temple University, Medical Education and Research Building, Room 1060, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Yuling Zhang
- Cardiovascular Medicine Department, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Yong Ji
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, China
| | - Jingfeng Wang
- Cardiovascular Medicine Department, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Xiao-Feng Yang
- Center for Metabolic Disease Research, Lewis Kats School of Medicine, Temple University, Medical Education and Research Building, Room 1060, 3500 N. Broad Street, Philadelphia, PA, 19140, USA.,Department of Pharmacology, Lewis Kats School of Medicine, Temple University, Philadelphia, PA, USA
| | - Hong Wang
- Center for Metabolic Disease Research, Lewis Kats School of Medicine, Temple University, Medical Education and Research Building, Room 1060, 3500 N. Broad Street, Philadelphia, PA, 19140, USA. .,Department of Pharmacology, Lewis Kats School of Medicine, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
32
|
Kimber I, Poole A, Basketter DA. Skin and respiratory chemical allergy: confluence and divergence in a hybrid adverse outcome pathway. Toxicol Res (Camb) 2018; 7:586-605. [PMID: 30090609 PMCID: PMC6060610 DOI: 10.1039/c7tx00272f] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 01/18/2018] [Indexed: 12/14/2022] Open
Abstract
Sensitisation of the respiratory tract to chemicals resulting in respiratory allergy and allergic asthma is an important occupational health problem, and presents toxicologists with no shortage of challenges. A major issue is that there are no validated or, even widely recognised, methods available for the identification and characterisation of chemical respiratory allergens, or for distinguishing respiratory allergens from contact allergens. The first objective here has been review what is known (and what is not known) of the mechanisms through which chemicals induce sensitisation of the respiratory tract, and to use this information to construct a hybrid Adverse Outcome Pathway (AOP) that combines consideration of both skin and respiratory sensitisation. The intention then has been to use the construction of this hybrid AOP to identify areas of commonality/confluence, and areas of departure/divergence, between skin sensitisation and sensitisation of the respiratory tract. The hybrid AOP not only provides a mechanistic understanding of how the processes of skin and respiratory sensitisation differ, buy also a means of identifying areas of uncertainty about chemical respiratory allergy that benefit from a further investment in research.
Collapse
Affiliation(s)
- Ian Kimber
- Faculty of Biology , Medicine and Health , University of Manchester , Oxford Road , Manchester M13 9PT , UK . ; Tel: +44 (0) 161 275 1587
| | - Alan Poole
- European Centre for Ecotoxicology and Toxicology of Chemicals (ECETOC) , 2 Av E Van Nieuwenhuyse , 1160 Brussels , Belgium
| | | |
Collapse
|
33
|
Takeda Y, Azuma M, Hatsugai R, Fujimoto Y, Hashimoto M, Fukase K, Matsumoto M, Seya T. The second and third amino acids of Pam2 lipopeptides are key for the proliferation of cytotoxic T cells. Innate Immun 2018; 24:323-331. [PMID: 29848176 PMCID: PMC6830919 DOI: 10.1177/1753425918777598] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The TLR2 agonist, dipalmitoyl lipopeptide (Pam2LP), has been used as an immune
adjuvant without much success. Pam2LP is recognised by TLR2/6 receptors in
humans and in mice. This study examined the proliferative activity of cytotoxic
T lymphocytes (CTL) using mouse Ag-presenting dendritic cells (DCs) and OT-I
assay system, where a library of synthetic Pam2LP was utilised from the
Staphylococcus aureus database. Ag-specific CTL expansion
and IFN-γ levels largely depended on the Pam2LP peptide sequence. The first Aa
is cysteine (Cys), which has an active SH residue to bridge fatty acids, and the
second and third Aa are hydrophilic or non-polar. The sequence structurally
adapted to the residual constitution of the reported TLR2/6 pocket. The inactive
sequence contained proline or leucine/isoleucine after the first Cys. Notably,
no direct activation of OT-I cells was detected without DCs by stimulation with
the active Pam2LP having the Cys-Ser sequence. MyD88, but not TICAM-1 or IFN
pathways, in DCs participates in DC maturation characterised by upregulation of
CD40, CD80 and CD86. Hence, the active Pam2LPs appear suitable for dimeric
TLR2/6 on DCs, resulting in induction of DC maturation.
Collapse
Affiliation(s)
- Yohei Takeda
- 1 Department of Vaccine Immunology, Graduate School of Medicine, Hokkaido University, Japan
| | - Masahiro Azuma
- 1 Department of Vaccine Immunology, Graduate School of Medicine, Hokkaido University, Japan
| | - Ryoko Hatsugai
- 1 Department of Vaccine Immunology, Graduate School of Medicine, Hokkaido University, Japan
| | - Yukari Fujimoto
- 2 Faculty of Science and Technology, Keio University, Japan.,3 Department of Chemistry, Graduate School of Science, Osaka University, Japan
| | - Masahito Hashimoto
- 4 Department of Nanostructure and Advanced Materials, Kagoshima University, Japan
| | - Koichi Fukase
- 3 Department of Chemistry, Graduate School of Science, Osaka University, Japan
| | - Misako Matsumoto
- 1 Department of Vaccine Immunology, Graduate School of Medicine, Hokkaido University, Japan
| | - Tsukasa Seya
- 1 Department of Vaccine Immunology, Graduate School of Medicine, Hokkaido University, Japan
| |
Collapse
|
34
|
Abstract
The immune system is characterized by the generation of structurally and functionally heterogeneous immune cells that constitute complex innate and adaptive immunity. This heterogeneity of immune cells results from changes in the expression of genes without altering DNA sequence. To achieve this heterogeneity, immune cells orchestrate the expression and functional status of transcription factor (TF) networks, which can be broadly categorized into 3 classes: pioneer TFs that facilitate initial commitment and differentiation of hematopoietic cells, subset-specific TFs that promote the generation of selected cell lineages, and immune-signaling TFs that regulate specialized function in differentiated cells. Epigenetic mechanisms are known to be critical for organizing the TF networks, thereby controlling immune cell lineage-fate decisions, plasticity, and function. The effects of epigenetic regulators can be heritable during cell mitosis, primarily through the modification of DNA and histone methylation patterns at gene loci. By doing so, the immune system is enabled to mount a selective but robust response to stimuli, such as pathogens, tumor cells, autoantigens, or allogeneic antigens in the setting of transplantation, while preserving the immune cell reservoir necessary for protecting the host against numerous other unexpected stimuli and limit detrimental effect of systemic inflammatory reactions.
Collapse
|
35
|
Effective Priming of Herpes Simplex Virus-Specific CD8 + T Cells In Vivo Does Not Require Infected Dendritic Cells. J Virol 2018; 92:JVI.01508-17. [PMID: 29142130 DOI: 10.1128/jvi.01508-17] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 11/06/2017] [Indexed: 11/20/2022] Open
Abstract
Resolution of virus infections depends on the priming of virus-specific CD8+ T cells by dendritic cells (DC). While this process requires major histocompatibility complex (MHC) class I-restricted antigen presentation by DC, the relative contribution to CD8+ T cell priming by infected DC is less clear. We have addressed this question in the context of a peripheral infection with herpes simplex virus 1 (HSV). Assessing the endogenous, polyclonal HSV-specific CD8+ T cell response, we found that effective in vivo T cell priming depended on the presence of DC subsets specialized in cross-presentation, while Langerhans cells and plasmacytoid DC were dispensable. Utilizing a novel mouse model that allows for the in vivo elimination of infected DC, we also demonstrated in vivo that this requirement for cross-presenting DC was not related to their infection but instead reflected their capacity to cross-present HSV-derived antigen. Taking the results together, this study shows that infected DC are not required for effective CD8+ T cell priming during a peripheral virus infection.IMPORTANCE The ability of some DC to present viral antigen to CD8+ T cells without being infected is thought to enable the host to induce killer T cells even when viruses evade or kill infected DC. However, direct experimental in vivo proof for this notion has remained elusive. The work described in this study characterizes the role that different DC play in the induction of virus-specific killer T cell responses and, critically, introduces a novel mouse model that allows for the selective elimination of infected DC in vivo Our finding that HSV-specific CD8+ T cells can be fully primed in the absence of DC infection shows that cross-presentation by DC is indeed sufficient for effective CD8+ T cell priming during a peripheral virus infection.
Collapse
|
36
|
Transcriptional mechanisms that control expression of the macrophage colony-stimulating factor receptor locus. Clin Sci (Lond) 2017; 131:2161-2182. [DOI: 10.1042/cs20170238] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/22/2017] [Accepted: 06/11/2017] [Indexed: 12/17/2022]
Abstract
The proliferation, differentiation, and survival of cells of the macrophage lineage depends upon signals from the macrophage colony-stimulating factor (CSF) receptor (CSF1R). CSF1R is expressed by embryonic macrophages and induced early in adult hematopoiesis, upon commitment of multipotent progenitors to the myeloid lineage. Transcriptional activation of CSF1R requires interaction between members of the E26 transformation-specific family of transcription factors (Ets) (notably PU.1), C/EBP, RUNX, AP-1/ATF, interferon regulatory factor (IRF), STAT, KLF, REL, FUS/TLS (fused in sarcoma/ranslocated in liposarcoma) families, and conserved regulatory elements within the mouse and human CSF1R locus. One element, the Fms-intronic regulatory element (FIRE), within intron 2, is conserved functionally across all the amniotes. Lineage commitment in multipotent progenitors also requires down-regulation of specific transcription factors such as MYB, FLI1, basic leucine zipper transcriptional factor ATF-like (BATF3), GATA-1, and PAX5 that contribute to differentiation of alternative lineages and repress CSF1R transcription. Many of these transcription factors regulate each other, interact at the protein level, and are themselves downstream targets of CSF1R signaling. Control of CSF1R transcription involves feed–forward and feedback signaling in which CSF1R is both a target and a participant; and dysregulation of CSF1R expression and/or function is associated with numerous pathological conditions. In this review, we describe the regulatory network behind CSF1R expression during differentiation and development of cells of the mononuclear phagocyte system.
Collapse
|
37
|
Burcelin R. [Gut microbiota and immune crosstalk in metabolic disease]. Biol Aujourdhui 2017; 211:1-18. [PMID: 28682223 DOI: 10.1051/jbio/2017008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Indexed: 05/28/2023]
Abstract
The aim of the review is to discuss about the role played by the defence crosstalk between the gut microbiota and the intestinal immune system, in the development of metabolic disease focusing on obesity and diabetes. Starting from physiological and pathological stand points and based on the latest published data, this review is addressing how the concept of the hologenome theory of evolution can drive the fate of metabolic disease. The notion of "metabolic infection" to explain the "metabolic inflammation" is discussed. This imply comments about the process of bacterial translocation and impaired intestinal immune defense against commensals. Eventually this review sets the soil for personalized medicine. The monthly increase in the number of publications on the gut microbiota to intestinal immune defense and the control of metabolism demonstrate the importance of this field of investigation. The notion of commensal as "self or non-self" has to be reevaluated in the light of the current data. Furthermore, data demonstrate the major role played by short chain fatty acids, secondary bile acids, LPS, peptidoglycans, indole derivatives, and other bacteria-related molecules on the shaping of cells involved in the intestinal protection against commensals is now becoming a central player in the incidence of metabolic diseases. The literature demonstrates that the onset of metabolic diseases and some specific co-morbidities can be explained by a gut microbiota to intestinal immune system crosstalk. Therefore, one should now consider this avenue of investigation as a putative source of biomarkers and therapeutic targets to personalize the treatment of metabolic disease and its co-morbidities. Gut microbiota is considered as a major regulator of metabolic disease. This reconciles the notion of metabolic inflammation and the epidemic development of the disease. In addition to evidence showing that a specific gut microbiota characterizes patients with obesity, type 2 diabetes, and hepatic steatosis, the mechanisms causal to the disease could be related to the translocation of microbiota from the gut to the tissues, which induces inflammation. The mechanisms regulating such a process are based on the crosstalk between the gut microbiota and the host immune system. The hologenome theory of evolution supports this concept and implies that therapeutic strategies aiming to control glycemia should take into account both the gut microbiota and the host immune system. This review discusses the latest evidence regarding the bidirectional impact of the gut microbiota on host immune system crosstalk for the control of metabolic disease, hyperglycemia, and obesity. To avoid redundancies with the literature, we will focus our attention on the intestinal immune system, identifying evidence for the generation of novel therapeutic strategies, which could be based on the control of the translocation of gut bacteria to tissues. Such novel strategies should hamper the role played by gut microbiota dysbiosis on the development of metabolic inflammation. Recent evidence in rodents allows us to conclude that an impaired intestinal immune system characterizes and could be causal in the development of metabolic disease. The fine understanding of the molecular mechanisms should allow for the development of a first line of treatment for metabolic disease and its co-morbidities.
Collapse
Affiliation(s)
- Rémy Burcelin
- Institut National de la Santé et de la Recherche Médicale (INSERM), 31024 Toulouse, France - Université Paul Sabatier (UPS), Unité Mixte de Recherche (UMR) 1048, Hôpital Rangueil, 31400 Toulouse, France - Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), 31432 Toulouse Cedex 4, France
| |
Collapse
|
38
|
Sichien D, Lambrecht BN, Guilliams M, Scott CL. Development of conventional dendritic cells: from common bone marrow progenitors to multiple subsets in peripheral tissues. Mucosal Immunol 2017; 10:831-844. [PMID: 28198365 DOI: 10.1038/mi.2017.8] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 01/14/2017] [Indexed: 02/04/2023]
Abstract
Our understanding of conventional dendritic cell (cDC) development and the functional specializations of distinct subsets in the peripheral tissues has increased greatly in recent years. Here, we review cDC development from the distinct progenitors in the bone marrow through to the distinct cDC subsets found in barrier tissues, providing an overview of the different subsets described in each location. In addition, we detail the transcription factors and local signals that have been proposed to control this developmental process. Importantly, despite these significant advances, numerous questions remain to be answered regarding cDC development. For example, it remains unclear whether the different subsets described, such as the CD103+CD11b+ and CD103-CD11b+ cDCs in the intestines, truly represent different populations or rather distinct developmental or activation stages. Furthermore, whether distinct progenitors exist for these cDC subsets remains to be determined. Thus in the last part of this review we discuss what we believe will be the main questions facing the field for the coming years.
Collapse
Affiliation(s)
- D Sichien
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - B N Lambrecht
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University, Ghent, Belgium
- Department of Pulmonary Medicine, ErasmusMC, Rotterdam, The Netherlands
| | - M Guilliams
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - C L Scott
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
39
|
Sun T, Rojas OL, Li C, Ward LA, Philpott DJ, Gommerman JL. Intestinal Batf3-dependent dendritic cells are required for optimal antiviral T-cell responses in adult and neonatal mice. Mucosal Immunol 2017; 10:775-788. [PMID: 27600308 DOI: 10.1038/mi.2016.79] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 07/27/2016] [Indexed: 02/06/2023]
Abstract
Although we know a great deal about which types of dendritic cells (DCs) promote T-cell priming in the periphery, less is known about which DC subset(s) provoke antiviral responses within the gut. Here we report that conventional Zbtb46-dependent DCs were critically required for antiviral CD8+ T-cell responses against rotavirus (RV), the major cause of childhood gastroenteritis worldwide. Furthermore, we found that in adult mice, Batf3-dependent DCs were required for generating optimal RV-specific CD8+ T-cell responses. However, in contrast to mice that lack Zbtb46-dependent DCs, a significant amount of interferon gamma-producing RV-specific CD8+ T cells were still detected in the small intestine of RV-infected adult Batf3-/- mice, suggesting the existence of compensatory cross-presentation mechanisms in the absence of Batf3-dependent DCs. In contrast to adult mice, we found that Batf3-dependent DCs were absolutely required for generating RV-specific CD8+ T-cell responses in neonates. Loss of Batf3-dependent DCs also resulted in a skewed polyclonal CD4+ T-cell response in both adult and neonatal mice upon RV infection, although local and systemic RV-specific immunoglobulin A production kinetics and titers were unimpaired. Our results provide insights that inform early-life vaccination strategies against RV infection.
Collapse
Affiliation(s)
- T Sun
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - O L Rojas
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - C Li
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - L A Ward
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - D J Philpott
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - J L Gommerman
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
40
|
|
41
|
Hypermethylation of Interferon Regulatory Factor 8 (IRF8) Confers Risk to Vogt-Koyanagi-Harada Disease. Sci Rep 2017; 7:1007. [PMID: 28432342 PMCID: PMC5430771 DOI: 10.1038/s41598-017-01249-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 03/27/2017] [Indexed: 12/15/2022] Open
Abstract
Aberrant methylation change of IRF8 confers risk to various tumors, and abnormal expression of IRF8 is involved in many autoimmune diseases, including ocular Behcet’s disease. However, whether the methylation change of IRF8 is associated with Vogt-Koyanagi-Harada (VKH) disease remains unknown. In the present study, we found a decreased IRF8 mRNA expression in association with a higher methylation level in monocyte-derived dendritic cells (DCs) from active VKH patients compared with the normal and inactive subjects. DCs incubated with cyclosporin a (CsA) or dexamethasone (DEX) showed a lower methylation and higher mRNA expression of IRF8 in active VKH patients. A demethylation reagent, 5-Aza-2′-deoxycytidine (DAC) showed a notable demethylation effect as evidenced by increasing the mRNA expression and reducing the methylation level of IRF8. It also suppressed the Th1 and Th17 responses through down-regulating the expression of co-stimulatory molecules (CD86, CD80, CD40), and reducing the production of pro-inflammatory cytokines (IL-6, IL-1β, IL-23, IL-12) produced by DCs. These findings shows that hypermethylation of IRF8 in DCs confers risk to VKH disease. Demethylation of IRF8 may offer a novel therapeutic strategy protect against VKH disease.
Collapse
|
42
|
Qiu Y, Zhu Y, Yu H, Yi S, Su W, Cao Q, Yuan G, Kijlstra A, Yang P. Ocular Behcet's disease is associated with aberrant methylation of interferon regulatory factor 8 (IRF8) in monocyte-derived dendritic cells. Oncotarget 2017; 8:51277-51287. [PMID: 28881647 PMCID: PMC5584248 DOI: 10.18632/oncotarget.17235] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 03/28/2017] [Indexed: 01/23/2023] Open
Abstract
Aberrant methylation of interferon regulatory factor 8 (IRF8) has been noted in various tumors. IRF8 has also been reported to be involved in many autoimmune diseases, including Behcet's disease (BD). However, the methylation status of IRF8 in BD has not been reported. To address this issue, we investigated whether the degree of methylation of IRF8 in dendritic cells (DCs) plays a role in the development of BD. We found a lower mRNA expression and a higher methylation level of IRF8 in active ocular BD patients as compared to normal subjects and inactive patients. Treatment with a demethylation agent, 5-Aza-2'-deoxycytidine (DAC) resulted in an increase of mRNA expression and a reduction of the IRF8 methylation level. It also down-regulated the expression of the co-stimulatory molecules CD86, CD80, CD40, and reduced the production of IL-6, IL-1β, IL-23 and IL-12. An inhibition of Th1/Th17 responses was observed as evidenced by a decreased production of IFN-γ, IL-17, and a reduction of IFN-γ/IL-17- producing CD4+ T cells following treatment with DAC. This study shows that active ocular BD patients have an aberrant IRF8 methylation status. These findings suggest that epigenetic control of IRF8 expression may offer a future target in the treatment of ocular BD.
Collapse
Affiliation(s)
- Yiguo Qiu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Yunyun Zhu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Hongsong Yu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Shenglan Yi
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Wencheng Su
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Qingfeng Cao
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Gangxiang Yuan
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Aize Kijlstra
- University Eye Clinic Maastricht, Maastricht, The Netherlands
| | - Peizeng Yang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| |
Collapse
|
43
|
Nasi A, Bollampalli VP, Sun M, Chen Y, Amu S, Nylén S, Eidsmo L, Rothfuchs AG, Réthi B. Immunogenicity is preferentially induced in sparse dendritic cell cultures. Sci Rep 2017; 7:43989. [PMID: 28276533 PMCID: PMC5343661 DOI: 10.1038/srep43989] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 02/02/2017] [Indexed: 12/16/2022] Open
Abstract
We have previously shown that human monocyte-derived dendritic cells (DCs) acquired different characteristics in dense or sparse cell cultures. Sparsity promoted the development of IL-12 producing migratory DCs, whereas dense cultures increased IL-10 production. Here we analysed whether the density-dependent endogenous breaks could modulate DC-based vaccines. Using murine bone marrow-derived DC models we show that sparse cultures were essential to achieve several key functions required for immunogenic DC vaccines, including mobility to draining lymph nodes, recruitment and massive proliferation of antigen-specific CD4+ T cells, in addition to their TH1 polarization. Transcription analyses confirmed higher commitment in sparse cultures towards T cell activation, whereas DCs obtained from dense cultures up-regulated immunosuppressive pathway components and genes suggesting higher differentiation plasticity towards osteoclasts. Interestingly, we detected a striking up-regulation of fatty acid and cholesterol biosynthesis pathways in sparse cultures, suggesting an important link between DC immunogenicity and lipid homeostasis regulation.
Collapse
Affiliation(s)
- Aikaterini Nasi
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | | | - Meng Sun
- Department of Medicine, Karolinska University Hospital and Karolinska Institutet, Solna, Sweden
| | - Yang Chen
- Department of Medicine, Science for Life Laboratory, Karolinska Institutet, Solna, Sweden
| | - Sylvie Amu
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Susanne Nylén
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Liv Eidsmo
- Department of Medicine, Karolinska University Hospital and Karolinska Institutet, Solna, Sweden
| | | | - Bence Réthi
- Department of Medicine, Karolinska University Hospital and Karolinska Institutet, Solna, Sweden
| |
Collapse
|
44
|
Abstract
The cDC1 subset of classical dendritic cells is specialized for priming CD8 T cell responses through the process of cross-presentation. The molecular mechanisms of cross-presentation remain incompletely understood because of limited biochemical analysis of rare cDC1 cells, difficulty in their genetic manipulation, and reliance on
in vitro systems based on monocyte- and bone-marrow-derived dendritic cells. This review will discuss cross-presentation from the perspective of studies with monocyte- or bone-marrow-derived dendritic cells while highlighting the need for future work examining cDC1 cells. We then discuss the role of cDC1s as a cellular platform to combine antigen processing for class I and class II MHC presentation to allow the integration of “help” from CD4 T cells during priming of CD8 T cell responses.
Collapse
Affiliation(s)
- Derek Theisen
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Kenneth Murphy
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Howard Hughes Medical Institute, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| |
Collapse
|
45
|
Chudnovskiy A, Mortha A, Kana V, Kennard A, Ramirez JD, Rahman A, Remark R, Mogno I, Ng R, Gnjatic S, Amir EAD, Solovyov A, Greenbaum B, Clemente J, Faith J, Belkaid Y, Grigg ME, Merad M. Host-Protozoan Interactions Protect from Mucosal Infections through Activation of the Inflammasome. Cell 2017; 167:444-456.e14. [PMID: 27716507 DOI: 10.1016/j.cell.2016.08.076] [Citation(s) in RCA: 234] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 07/05/2016] [Accepted: 08/30/2016] [Indexed: 12/23/2022]
Abstract
While conventional pathogenic protists have been extensively studied, there is an underappreciated constitutive protist microbiota that is an integral part of the vertebrate microbiome. The impact of these species on the host and their potential contributions to mucosal immune homeostasis remain poorly studied. Here, we show that the protozoan Tritrichomonas musculis activates the host epithelial inflammasome to induce IL-18 release. Epithelial-derived IL-18 promotes dendritic cell-driven Th1 and Th17 immunity and confers dramatic protection from mucosal bacterial infections. Along with its role as a "protistic" antibiotic, colonization with T. musculis exacerbates the development of T-cell-driven colitis and sporadic colorectal tumors. Our findings demonstrate a novel mutualistic host-protozoan interaction that increases mucosal host defenses at the cost of an increased risk of inflammatory disease.
Collapse
Affiliation(s)
- Aleksey Chudnovskiy
- Department of Oncological Science, Icahn School of Medicine at Mount Sinai, 1475 Madison Avenue, New York, NY 10028, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1475 Madison Avenue, New York, NY 10028, USA; The Immunological Institute, Icahn School of Medicine at Mount Sinai, 1475 Madison Avenue, New York, NY 10028, USA
| | - Arthur Mortha
- Department of Oncological Science, Icahn School of Medicine at Mount Sinai, 1475 Madison Avenue, New York, NY 10028, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1475 Madison Avenue, New York, NY 10028, USA; The Immunological Institute, Icahn School of Medicine at Mount Sinai, 1475 Madison Avenue, New York, NY 10028, USA
| | - Veronika Kana
- Department of Oncological Science, Icahn School of Medicine at Mount Sinai, 1475 Madison Avenue, New York, NY 10028, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1475 Madison Avenue, New York, NY 10028, USA; The Immunological Institute, Icahn School of Medicine at Mount Sinai, 1475 Madison Avenue, New York, NY 10028, USA
| | - Andrea Kennard
- Molecular Parasitology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Juan David Ramirez
- Grupo de Investigaciones Microbiologicas-UR (GIMUR), Universidad del Rosario, Bogotá, Colombia; Molecular Parasitology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Adeeb Rahman
- The Immunological Institute, Icahn School of Medicine at Mount Sinai, 1475 Madison Avenue, New York, NY 10028, USA; Department of Genetics & Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, 1475 Madison Avenue, New York, NY 10028, USA
| | - Romain Remark
- Department of Oncological Science, Icahn School of Medicine at Mount Sinai, 1475 Madison Avenue, New York, NY 10028, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1475 Madison Avenue, New York, NY 10028, USA; The Immunological Institute, Icahn School of Medicine at Mount Sinai, 1475 Madison Avenue, New York, NY 10028, USA
| | - Ilaria Mogno
- Department of Genetics & Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, 1475 Madison Avenue, New York, NY 10028, USA
| | - Ruby Ng
- The Immunological Institute, Icahn School of Medicine at Mount Sinai, 1475 Madison Avenue, New York, NY 10028, USA
| | - Sasha Gnjatic
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1475 Madison Avenue, New York, NY 10028, USA; The Immunological Institute, Icahn School of Medicine at Mount Sinai, 1475 Madison Avenue, New York, NY 10028, USA
| | - El-Ad David Amir
- Department of Oncological Science, Icahn School of Medicine at Mount Sinai, 1475 Madison Avenue, New York, NY 10028, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1475 Madison Avenue, New York, NY 10028, USA; The Immunological Institute, Icahn School of Medicine at Mount Sinai, 1475 Madison Avenue, New York, NY 10028, USA
| | - Alexander Solovyov
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1475 Madison Avenue, New York, NY 10028, USA
| | - Benjamin Greenbaum
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1475 Madison Avenue, New York, NY 10028, USA
| | - Jose Clemente
- The Immunological Institute, Icahn School of Medicine at Mount Sinai, 1475 Madison Avenue, New York, NY 10028, USA; Department of Genetics & Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, 1475 Madison Avenue, New York, NY 10028, USA
| | - Jeremiah Faith
- The Immunological Institute, Icahn School of Medicine at Mount Sinai, 1475 Madison Avenue, New York, NY 10028, USA; Department of Genetics & Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, 1475 Madison Avenue, New York, NY 10028, USA
| | - Yasmine Belkaid
- Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA; NIAID Microbiome Program, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Michael E Grigg
- Molecular Parasitology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Miriam Merad
- Department of Oncological Science, Icahn School of Medicine at Mount Sinai, 1475 Madison Avenue, New York, NY 10028, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1475 Madison Avenue, New York, NY 10028, USA; The Immunological Institute, Icahn School of Medicine at Mount Sinai, 1475 Madison Avenue, New York, NY 10028, USA.
| |
Collapse
|
46
|
Abstract
The discovery of long noncoding RNAs (lncRNA) has provided a new perspective on gene regulation in diverse biological contexts. lncRNAs are remarkably versatile molecules that interact with RNA, DNA, or proteins to promote or restrain the expression of protein-coding genes. Activation of immune cells is associated with dynamic changes in expression of genes, the products of which combat infectious microorganisms, initiate repair, and resolve inflammatory responses in cells and tissues. Recent evidence indicates that lncRNAs play important roles in directing the development of diverse immune cells and controlling the dynamic transcriptional programs that are a hallmark of immune cell activation. The importance of these molecules is underscored by their newly recognized roles in inflammatory diseases. In this review, we discuss the contribution of lncRNAs in the development and activation of immune cells and their roles in immune-related diseases. We also discuss challenges faced in identifying biological functions for this large and complex class of genes.
Collapse
Affiliation(s)
- Maninjay K Atianand
- Program in Innate Immunity, Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605;
| | - Daniel R Caffrey
- Program in Innate Immunity, Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605;
| | - Katherine A Fitzgerald
- Program in Innate Immunity, Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605;
| |
Collapse
|
47
|
Batf3 selectively determines acquisition of CD8 + dendritic cell phenotype and function. Immunol Cell Biol 2016; 95:215-223. [PMID: 27897162 PMCID: PMC5309136 DOI: 10.1038/icb.2016.83] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 08/29/2016] [Accepted: 08/30/2016] [Indexed: 12/26/2022]
Abstract
Batf3 is a transcription factor that impacts the development of CD103+ tissue-resident dendritic cells (DCs). However, whether Batf3 is absolutely required for the development of CD8+ DCs remains controversial. Id2 is required for CD8+ DC development. Here we show that bone marrow chimeric mice with a deletion of Id2 in the CD11c compartment lose the ability to reject a skin graft expressing a non-self protein antigen or mount a delayed hypersensitivity response. In contrast, Batf3-/- mice remained competent for skin graft rejection and delayed hypersensitivity, and retained a CD8+ DC population with markers characteristic of the CD11b+ DC lineage, including CD11b, CD4 and CD172α, as well as the key regulator transcription factor IRF4, but lacked IRF8 expression. CD8+ DCs in Batf3-/- mice took up and cleaved protein antigen and larger particles but were unable to phagocytose dying cells, a characteristic feature to the CD8+ DC lineage. These data clarify a requirement for CD8+ lineage DCs to induce effectors of neo-antigen-driven skin graft rejection, and improve our understanding of DC subtype commitment by demonstrating that in the absence of Batf3 CD8+ DCs can change their fate and become CD11b+ DCs.
Collapse
|
48
|
Malloy AMW, Ruckwardt TJ, Morabito KM, Lau-Kilby AW, Graham BS. Pulmonary Dendritic Cell Subsets Shape the Respiratory Syncytial Virus-Specific CD8+ T Cell Immunodominance Hierarchy in Neonates. THE JOURNAL OF IMMUNOLOGY 2016; 198:394-403. [PMID: 27895172 DOI: 10.4049/jimmunol.1600486] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 10/31/2016] [Indexed: 01/25/2023]
Abstract
Young infants are generally more susceptible to viral infections and experience more severe disease than do adults. CD8+ T cells are important for viral clearance, and although often ineffective in neonates they can be protective when adequately stimulated. Using a murine CB6F1/J hybrid model of respiratory syncytial virus (RSV) infection, we previously demonstrated that the CD8+ T cell immunodominance hierarchy to two RSV-derived epitopes, KdM282-90 and DbM187-195, was determined by the age at infection. To determine whether age-dependent RSV-specific CD8+ T cell responses could be modified through enhanced innate signaling, we used TLR4 or TLR9 agonist treatment at the time of infection, which remarkably changed the neonatal codominant response to an adult-like KdM282-90 CD8+ T cell immunodominant response. This shift was associated with an increase in the number of conventional dendritic cells, CD11b+ and CD103+ dendritic cells, in the lung-draining lymph node, as well as increased expression of the costimulatory molecule CD86. The magnitude of the KdM282-90 CD8+ T cell response in TLR agonist-treated neonates could be blocked with Abs against CD80 and CD86. These studies demonstrate the age-dependent function of conventional dendritic cells, their role in determining immunodominance hierarchy, and epitope-specific CD8+ T cell requirements for costimulation, all of which influence the immune response magnitude. The unique impact of TLR agonists on neonatal T cell responses is important to consider for RSV vaccines designed for young infants.
Collapse
Affiliation(s)
- Allison M W Malloy
- Laboratory of Neonatal Infection and Immunity, Department of Pediatrics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814; and .,Viral Pathogenesis Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Tracy J Ruckwardt
- Viral Pathogenesis Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Kaitlyn M Morabito
- Viral Pathogenesis Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Annie W Lau-Kilby
- Laboratory of Neonatal Infection and Immunity, Department of Pediatrics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814; and
| | - Barney S Graham
- Viral Pathogenesis Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
49
|
Scheenstra MR, De Cuyper IM, Branco-Madeira F, de Bleser P, Kool M, Meinders M, Hoogenboezem M, Mul E, Wolkers MC, Salerno F, Nota B, Saeys Y, Klarenbeek S, van IJcken WFJ, Hammad H, Philipsen S, van den Berg TK, Kuijpers TW, Lambrecht BN, Gutiérrez L. GATA1-Deficient Dendritic Cells Display Impaired CCL21-Dependent Migration toward Lymph Nodes Due to Reduced Levels of Polysialic Acid. THE JOURNAL OF IMMUNOLOGY 2016; 197:4312-4324. [DOI: 10.4049/jimmunol.1600103] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 09/29/2016] [Indexed: 02/05/2023]
|
50
|
Lund H, Boysen P, Åkesson CP, Lewandowska-Sabat AM, Storset AK. Transient Migration of Large Numbers of CD14(++) CD16(+) Monocytes to the Draining Lymph Node after Onset of Inflammation. Front Immunol 2016; 7:322. [PMID: 27621730 PMCID: PMC5002921 DOI: 10.3389/fimmu.2016.00322] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 08/15/2016] [Indexed: 12/12/2022] Open
Abstract
The dynamics of skin-draining cells following infection or vaccination provide important insight into the initiation of immune responses. In this study, the local recruitment and activation of immune cells in draining lymph nodes (LNs) was studied in calves in an adjuvant-induced inflammation. A transient but remarkably strong recruitment of monocytes was demonstrated after onset of inflammation, constituting up to 41% of live cells in the draining LNs after 24 h. Numerous CD14(+) cells were visualized in subcutaneous tissues and draining LNs, and the majority of these cells did not express dendritic cell-associated markers CD205 and CD11c. In the LNs, recruited cells were predominately of a CD14(++) and CD16(+) phenotype, consistent with an intermediate monocyte subset characterized to possess a high inflammatory potential. Moreover, monocytes from the draining LN showed a high expression of genes coding for pro-inflammatory cytokines, including IL-1β, IL-6, TNFa, and TGFβ. Shortly after their appearance in the LN cortical areas, the monocytes had moved into the medulla followed by an increase in peripheral blood. In conclusion, this study provides novel information on in vivo monocyte recruitment and migration after onset of inflammation.
Collapse
Affiliation(s)
- Hege Lund
- Department of Food Safety and Infection Biology, Norwegian University of Life Sciences , Oslo , Norway
| | - Preben Boysen
- Department of Food Safety and Infection Biology, Norwegian University of Life Sciences , Oslo , Norway
| | - Caroline Piercey Åkesson
- Department of Basic Sciences and Aquatic Medicine, Norwegian University of Life Sciences , Oslo , Norway
| | | | - Anne K Storset
- Department of Food Safety and Infection Biology, Norwegian University of Life Sciences , Oslo , Norway
| |
Collapse
|