38 results on '"Carlos G. Briseño"'
Search Results
2. Distinct Transcriptional Programs Control Cross-Priming in Classical and Monocyte-Derived Dendritic Cells
- Author
-
Carlos G. Briseño, Malay Haldar, Nicole M. Kretzer, Xiaodi Wu, Derek J. Theisen, Wumesh KC, Vivek Durai, Gary E. Grajales-Reyes, Arifumi Iwata, Prachi Bagadia, Theresa L. Murphy, and Kenneth M. Murphy
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Both classical DCs (cDCs) and monocyte-derived DCs (Mo-DCs) are capable of cross-priming CD8+ T cells in response to cell-associated antigens. We found that Ly-6ChiTREML4− monocytes can differentiate into Zbtb46+ Mo-DCs in response to granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-4 (IL-4) but that Ly-6ChiTREML4+ monocytes were committed to differentiate into Ly-6CloTREML4+ monocytes. Differentiation of Zbtb46+ Mo-DCs capable of efficient cross-priming required both GM-CSF and IL-4 and was accompanied by the induction of Batf3 and Irf4. However, monocytes require IRF4, but not BATF3, to differentiate into Zbtb46+ Mo-DCs capable of cross-priming CD8+ T cells. Instead, Irf4−/− monocytes differentiate into macrophages in response to GM-CSF and IL-4. Thus, cDCs and Mo-DCs require distinct transcriptional programs of differentiation in acquiring the capacity to prime CD8+ T cells. These differences may be of consideration in the use of therapeutic DC vaccines based on Mo-DCs.
- Published
- 2016
- Full Text
- View/download PDF
3. Oral Antibiotic Treatment of Mice Exacerbates the Disease Severity of Multiple Flavivirus Infections
- Author
-
Larissa B. Thackray, Scott A. Handley, Matthew J. Gorman, Subhajit Poddar, Prachi Bagadia, Carlos G. Briseño, Derek J. Theisen, Qing Tan, Barry L. Hykes, Jr., Hueylie Lin, Tiffany M. Lucas, Chandni Desai, Jeffrey I. Gordon, Kenneth M. Murphy, Herbert W. Virgin, and Michael S. Diamond
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: Although the outcome of flavivirus infection can vary from asymptomatic to lethal, environmental factors modulating disease severity are poorly defined. Here, we observed increased susceptibility of mice to severe West Nile (WNV), Dengue, and Zika virus infections after treatment with oral antibiotics (Abx) that depleted the gut microbiota. Abx treatment impaired the development of optimal T cell responses, with decreased levels of WNV-specific CD8+ T cells associated with increased infection and immunopathology. Abx treatments that resulted in enhanced WNV susceptibility generated changes in the overall structure of the gut bacterial community and in the abundance of specific bacterial taxa. As little as 3 days of treatment with ampicillin was sufficient to alter host immunity and WNV outcome. Our results identify oral Abx therapy as a potential environmental determinant of systemic viral disease, and they raise the possibility that perturbation of the gut microbiota may have deleterious consequences for subsequent flavivirus infections. : Thackray et al. observed increased susceptibility to West Nile, Zika, and Dengue virus infections following oral antibiotic treatment in mice. Antibiotics altered the bacterial abundance and community structure and the development of optimal T cell immunity. These data suggest that antibiotics may have deleterious consequences for subsequent flavivirus infections. Keywords: West Nile virus, Dengue virus, Zika virus, flavivirus, oral antibiotics, gut microbiota, risk factors, pathogenesis determinants, immunity
- Published
- 2018
- Full Text
- View/download PDF
4. Correction: Corrigendum: Regulation of monocyte cell fate by blood vessels mediated by Notch signalling
- Author
-
Jaba Gamrekelashvili, Roberto Giagnorio, Jasmin Jussofie, Oliver Soehnlein, Johan Duchene, Carlos G. Briseño, Saravana K. Ramasamy, Kashyap Krishnasamy, Anne Limbourg, Christine Häger, Tamar Kapanadze, Chieko Ishifune, Rabea Hinkel, Freddy Radtke, Lothar J. Strobl, Ursula Zimber-Strobl, L. Christian Napp, Johann Bauersachs, Hermann Haller, Koji Yasutomo, Christian Kupatt, Kenneth M. Murphy, Ralf H. Adams, Christian Weber, and Florian P. Limbourg
- Subjects
Science - Abstract
Nature Communications 7: Article number: 12597 (2016); Published: 31 August 2016; Updated: 3 May 2017 The authors inadvertently omitted Christine Häger, who was involved in the initial characterization of Notch mutant mice presented in this Article, from the author list and Author contributions statement.
- Published
- 2017
- Full Text
- View/download PDF
5. Phagocytosis increases an oxidative metabolic and immune suppressive signature in tumor macrophages
- Author
-
Michael A. Gonzalez, Daniel R. Lu, Maryam Yousefi, Ashley Kroll, Chen Hao Lo, Carlos G. Briseño, J. E. Vivienne Watson, Sergey Novitskiy, Vanessa Arias, Hong Zhou, Andres Plata Stapper, Min K. Tsai, Emily L. Ashkin, Christopher W. Murray, Chi-Ming Li, Monte M. Winslow, and Kristin V. Tarbell
- Subjects
Immunology ,Immunology and Allergy - Abstract
Phagocytosis is a key macrophage function, but how phagocytosis shapes tumor-associated macrophage (TAM) phenotypes and heterogeneity in solid tumors remains unclear. Here, we utilized both syngeneic and novel autochthonous lung tumor models in which neoplastic cells express the fluorophore tdTomato (tdTom) to identify TAMs that have phagocytosed neoplastic cells in vivo. Phagocytic tdTompos TAMs upregulated antigen presentation and anti-inflammatory proteins, but downregulated classic proinflammatory effectors compared to tdTomneg TAMs. Single-cell transcriptomic profiling identified TAM subset-specific and common gene expression changes associated with phagocytosis. We uncover a phagocytic signature that is predominated by oxidative phosphorylation (OXPHOS), ribosomal, and metabolic genes, and this signature correlates with worse clinical outcome in human lung cancer. Expression of OXPHOS proteins, mitochondrial content, and functional utilization of OXPHOS were increased in tdTompos TAMs. tdTompos tumor dendritic cells also display similar metabolic changes. Our identification of phagocytic TAMs as a distinct myeloid cell state links phagocytosis of neoplastic cells in vivo with OXPHOS and tumor-promoting phenotypes.
- Published
- 2023
- Full Text
- View/download PDF
6. cDC1 prime and are licensed by CD4+ T cells to induce anti-tumour immunity
- Author
-
Vivek Durai, Tiantian Liu, Wayne M. Yokoyama, Prachi Bagadia, Theresa L. Murphy, Derek J. Theisen, Jesse T. Davidson, Jeffrey P. Ward, Renee Wu, Stephen T. Ferris, Lijin Li, William E. Gillanders, Carlos G. Briseño, Michael D. Bern, Robert D. Schreiber, Kenneth M. Murphy, and Gregory F. Wu
- Subjects
0301 basic medicine ,Multidisciplinary ,CD40 ,biology ,Chemistry ,Antigen processing ,T cell ,Priming (immunology) ,Article ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Antigen ,Immunity ,030220 oncology & carcinogenesis ,biology.protein ,medicine ,Signal transduction ,CD8 - Abstract
Conventional type 1 dendritic cells (cDC1)1 are thought to perform antigen cross-presentation, which is required to prime CD8+ T cells2,3, whereas cDC2 are specialized for priming CD4+ T cells4,5. CD4+ T cells are also considered to help CD8+ T cell responses through a variety of mechanisms6–11, including a process whereby CD4+ T cells ‘license’ cDC1 for CD8+ T cell priming12. However, this model has not been directly tested in vivo or in the setting of help-dependent tumour rejection. Here we generated an Xcr1Cre mouse strain to evaluate the cellular interactions that mediate tumour rejection in a model requiring CD4+ and CD8+ T cells. As expected, tumour rejection required cDC1 and CD8+ T cell priming required the expression of major histocompatibility class I molecules by cDC1. Unexpectedly, early priming of CD4+ T cells against tumour-derived antigens also required cDC1, and this was not simply because they transport antigens to lymph nodes for processing by cDC2, as selective deletion of major histocompatibility class II molecules in cDC1 also prevented early CD4+ T cell priming. Furthermore, deletion of either major histocompatibility class II or CD40 in cDC1 impaired tumour rejection, consistent with a role for cognate CD4+ T cell interactions and CD40 signalling in cDC1 licensing. Finally, CD40 signalling in cDC1 was critical not only for CD8+ T cell priming, but also for initial CD4+ T cell activation. Thus, in the setting of tumour-derived antigens, cDC1 function as an autonomous platform capable of antigen processing and priming for both CD4+ and CD8+ T cells and of the direct orchestration of their cross-talk that is required for optimal anti-tumour immunity. Conventional type 1 dendritic cells perform antigen processing and priming of CD4+ and CD8+ T cells against tumour antigens, orchestrating their cross-talk to effect anti-tumour immunity.
- Published
- 2020
- Full Text
- View/download PDF
7. Indoleamine 2,3-dioxygenase 1 activation in mature cDC1 promotes tolerogenic education of inflammatory cDC2 via metabolic communication
- Author
-
Marco Gargaro, Giulia Scalisi, Giorgia Manni, Carlos G. Briseño, Prachi Bagadia, Vivek Durai, Derek J. Theisen, Sunkyung Kim, Marilena Castelli, Chenling A. Xu, Gerd Meyer zu Hörste, Giuseppe Servillo, Maria A. Della Fazia, Giulia Mencarelli, Doriana Ricciuti, Eleonora Padiglioni, Nicola Giacchè, Carolina Colliva, Roberto Pellicciari, Mario Calvitti, Teresa Zelante, Dietmar Fuchs, Ciriana Orabona, Louis Boon, Alban Bessede, Marco Colonna, Paolo Puccetti, Theresa L. Murphy, Kenneth M. Murphy, and Francesca Fallarino
- Subjects
IL-6 ,Immunology ,AhR ,Tryptophan ,RelB ,Dendritic Cells ,Indoleamine-Pyrrole 2 ,kynurenine ,neuroinflammation ,Mice ,Infectious Diseases ,IDO1 ,Dioxygenase ,Immunology and Allergy ,Animals ,Humans ,Indoleamine-Pyrrole 2,3,-Dioxygenase ,tryptophan metabolism ,metabolites ,immunotolerance ,Signal Transduction - Abstract
Conventional dendritic cells (cDCs), cDC1 and cDC2, act both to initiate immunity and maintain self-tolerance. The tryptophan metabolic enzyme indoleamine 2,3-dioxygenase 1 (IDO1) is used by cDCs in maintaining tolerance, but its role in different subsets remains unclear. At homeostasis, only mature CCR7sup+/supcDC1 expressed IDO1 that was dependent on IRF8. Lipopolysaccharide treatment induced maturation and IDO1-dependent tolerogenic activity in isolated immature cDC1, but not isolated cDC2. However, both human and mouse cDC2 could induce IDO1 and acquire tolerogenic function when co-cultured with mature cDC1 through the action of cDC1-derived l-kynurenine. Accordingly, cDC1-specific inactivation of IDO1 in vivo exacerbated disease in experimental autoimmune encephalomyelitis. This study identifies a previously unrecognized metabolic communication in which IDO1-expressing cDC1 cells extend their immunoregulatory capacity to the cDC2 subset through their production of tryptophan metabolite l-kynurenine. This metabolic axis represents a potential therapeutic target in treating autoimmune demyelinating diseases.
- Published
- 2022
8. Cryptic activation of an Irf8 enhancer governs cDC1 fate specification
- Author
-
Kenneth M. Murphy, Gary E. Grajales-Reyes, Rodney D. Newberry, Carlos G. Briseño, Prachi Bagadia, Howard Y. Chang, Theresa L. Murphy, Vivek Durai, Swapneel J. Patel, Xiao Huang, Hiromi Tagoh, Jeffrey M. Granja, Miriam Wöhner, Jesse T. Davidson, Barbara L. Kee, Ansuman T. Satpathy, Tian Tian Liu, Renee Wu, Meinrad Busslinger, Arifumi Iwata, and Devesha H. Kulkarni
- Subjects
0301 basic medicine ,Cellular differentiation ,Immunology ,Biology ,Article ,Monocytes ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Tumor Cells, Cultured ,Immunology and Allergy ,Animals ,Cell Lineage ,Progenitor cell ,Enhancer ,Transcription factor ,Regulation of gene expression ,Mice, Knockout ,Macrophages ,Stem Cells ,Cell Differentiation ,Dendritic cell ,Dendritic Cells ,Cell biology ,Chromatin ,Mice, Inbred C57BL ,030104 developmental biology ,Enhancer Elements, Genetic ,Gene Expression Regulation ,Interferon Regulatory Factors ,IRF8 ,CRISPR-Cas Systems ,030215 immunology - Abstract
Induction of the transcription factor Irf8 in the common dendritic cell progenitor (CDP) is required for classical type 1 dendritic cell (cDC1) fate specification, but the mechanisms controlling this induction are unclear. In the present study Irf8 enhancers were identified via chromatin profiling of dendritic cells and CRISPR/Cas9 genome editing was used to assess their roles in Irf8 regulation. An enhancer 32 kilobases (kb) downstream of the Irf8 transcriptional start site (+32-kb Irf8) that was active in mature cDC1s was required for the development of this lineage, but not for its specification. Instead, a +41-kb Irf8 enhancer, previously thought to be active only in plasmacytoid dendritic cells, was found to also be transiently accessible in cDC1 progenitors, and deleting this enhancer prevented the induction of Irf8 in CDPs and abolished cDC1 specification. Thus, cryptic activation of the +41-kb Irf8 enhancer in dendritic cell progenitors is responsible for cDC1 fate specification. The transcription factor IRF8 is essential for classical type 1 dendritic cell (cDC1) development. Murphy and colleagues investigate in detail the molecular control of cDC1 fate specification by systematically unpicking the IRF8 enhancer regions.
- Published
- 2019
9. Batf3-Dependent Genes Control Tumor Rejection Induced by Dendritic Cells Independently of Cross-Presentation
- Author
-
Kenneth M. Murphy, Carlos G. Briseño, Nicole M. Kretzer, Stephen T. Ferris, Derek J. Theisen, Arifumi Iwata, and Theresa L. Murphy
- Subjects
0301 basic medicine ,Cancer Research ,Lineage (genetic) ,Transgene ,Immunology ,Cross-presentation ,Biology ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,Antigen ,Cell culture ,IRF8 ,Progenitor cell ,CD8 - Abstract
The BATF3-dependent cDC1 lineage of conventional dendritic cells (cDC) is required for rejection of immunogenic sarcomas and for rejection of progressive sarcomas during checkpoint blockade therapy. One unique function of the cDC1 lineage is the efficient cross-presentation of tumor-derived neoantigens to CD8+ T cells, but it is not clear that this is the only unique function of cDC1 required for tumor rejection. We previously showed that BATF3 functions during cDC1 lineage commitment to maintain IRF8 expression in the specified cDC1 progenitor. However, since cDC1 progenitors do not develop into mature cDC1s in Batf3−/− mice, it is still unclear whether BATF3 has additional functions in mature cDC1 cells. A transgenic Irf8-Venus reporter allele increases IRF8 protein concentration sufficiently to allow autonomous cDC1 development in spleens of Batf3−/− mice. These restored Batf3−/− cDC1s are transcriptionally similar to control wild-type cDC1s but have reduced expression of a restricted set of cDC1-specific genes. Restored Batf3−/− cDC1s are able to cross-present cell-associated antigens both in vitro and in vivo. However, Batf3−/− cDC1 exhibit altered characteristics in vivo and are unable to mediate tumor rejection. These results show that BATF3, in addition to regulating Irf8 expression to stabilize cDC1 lineage commitment, also controls expression of a small set of genes required for cDC1-mediated tumor rejection. These BATF3-regulated genes may be useful targets in immunotherapies aimed at promoting tumor rejection.
- Published
- 2019
- Full Text
- View/download PDF
10. WDFY4 is required for cross-presentation in response to viral and tumor antigens
- Author
-
Derek J. Theisen, Wayne M. Yokoyama, Vivek Durai, Wandy L. Beatty, Nima Mosammaparast, Marco Gargaro, Herbert W. Virgin, William E. Gillanders, Robert D. Schreiber, Carlos G. Briseño, Qiuling Wang, Theresa L. Murphy, Joshua R. Brickner, Prachi Bagadia, L. David Sibley, Michael S. Diamond, Kenneth M. Murphy, Jesse T. Davidson, Pritesh Desai, and Elvin J. Lauron
- Subjects
0301 basic medicine ,XCR1 ,CD8-Positive T-Lymphocytes ,Mice ,03 medical and health sciences ,Cross-Priming ,0302 clinical medicine ,Antigen ,Antigens, Neoplasm ,In vivo ,Animals ,Humans ,CRISPR ,Genetic Testing ,Antigens, Viral ,Multidisciplinary ,biology ,Intracellular Signaling Peptides and Proteins ,Toxoplasma gondii ,Cross-presentation ,biology.organism_classification ,Mice, Mutant Strains ,Mice, Inbred C57BL ,Repressor Proteins ,Basic-Leucine Zipper Transcription Factors ,030104 developmental biology ,030220 oncology & carcinogenesis ,Tumor rejection ,Immunology ,CRISPR-Cas Systems ,Toxoplasma ,Toxoplasmosis ,CD8 - Abstract
Adding to the cross-presentation family Immune responses to viral or tumor antigens are typically initiated by the process of cross-presentation. Cross-presentation is believed to be the major way that innate immune cells, such as the classical dendritic cell 1 (cDC1) subset, activate and prime immunological T cells. Theisen et al. used CRISPR-based screening to identify regulators of cross-presentation by cDC1s (see the Perspective by Barbet and Blander). One such regulator that was identified, WDFY4 (WD repeat- and FYVE domain–containing protein 4), was required for cross-presentation of cell- and bacterial-associated antigens. WDFY4 played a critical role in cDC1-mediated viral and tumor immunity yet did not seem necessary for major histocompatibility complex class II presentation or for cross-presentation by monocyte-derived DCs. Science , this issue p. 694 ; see also p. 641
- Published
- 2018
- Full Text
- View/download PDF
11. Notch2-dependent DC2s mediate splenic germinal center responses
- Author
-
Kevin W. O'Connor, Theresa L. Murphy, Vivek Durai, Jesse T. Davidson, Kenneth M. Murphy, Derek J. Theisen, Stephen T. Ferris, Prachi Bagadia, Carlos G. Briseño, and Ansuman T. Satpathy
- Subjects
0301 basic medicine ,endocrine system ,Erythrocytes ,dendritic cell ,Lymphocyte Activation ,Kruppel-Like Factor 4 ,Mice ,03 medical and health sciences ,Immunology and Inflammation ,0302 clinical medicine ,Immune system ,Antigen ,Genetic model ,MHC class I ,medicine ,Animals ,Receptor, Notch2 ,T follicular helper cell ,Cells, Cultured ,B cell ,Mice, Knockout ,Antigen Presentation ,B-Lymphocytes ,Sheep ,Multidisciplinary ,biology ,Germinal center ,Cell Differentiation ,Dendritic Cells ,T-Lymphocytes, Helper-Inducer ,Dendritic cell ,Biological Sciences ,Germinal Center ,Immunity, Humoral ,3. Good health ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Humoral immunity ,biology.protein ,Spleen ,Signal Transduction ,030215 immunology - Abstract
Significance High-affinity antibody responses involve selection of B cells in the germinal center (GC) by cognate interactions with T follicular helper (TFH) cells, which in turn must first be activated by classical dendritic cells (cDCs). We observe that Notch2-dependent cDC2s are required in vivo for induction of TFH cells, GC B cells, and specific antibody production in response to sheep red blood cell (SRBC) immunization. Notch2 signaling impacted a broad transcriptional program in cDC2s both at homeostasis and after SRBC immunization, although we have not identified a target gene that mediates TFH differentiation. Thus, Notch2 is a transcription factor that acts in cDCs and is selectively required for support of the GC reaction., CD4+ T follicular helper (TFH) cells support germinal center (GC) reactions promoting humoral immunity. Dendritic cell (DC) diversification into genetically distinct subsets allows for specialization in promoting responses against several types of pathogens. Whether any classical DC (cDC) subset is required for humoral immunity is unknown, however. We tested several genetic models that selectively ablate distinct DC subsets in mice for their impact on splenic GC reactions. We identified a requirement for Notch2-dependent cDC2s, but not Batf3-dependent cDC1s or Klf4-dependent cDC2s, in promoting TFH and GC B cell formation in response to sheep red blood cells and inactivated Listeria monocytogenes. This effect was mediated independent of Il2ra and several Notch2-dependent genes expressed in cDC2s, including Stat4 and Havcr2. Notch2 signaling during cDC2 development also substantially reduced the efficiency of cDC2s for presentation of MHC class II-restricted antigens, limiting the strength of CD4 T cell activation. Together, these results demonstrate a nonredundant role for the Notch2-dependent cDC2 subset in supporting humoral immune responses.
- Published
- 2018
- Full Text
- View/download PDF
12. cDC1 prime and are licensed by CD4
- Author
-
Stephen T, Ferris, Vivek, Durai, Renee, Wu, Derek J, Theisen, Jeffrey P, Ward, Michael D, Bern, Jesse T, Davidson, Prachi, Bagadia, Tiantian, Liu, Carlos G, Briseño, Lijin, Li, William E, Gillanders, Gregory F, Wu, Wayne M, Yokoyama, Theresa L, Murphy, Robert D, Schreiber, and Kenneth M, Murphy
- Subjects
CD4-Positive T-Lymphocytes ,Antigen Presentation ,Mice ,Cross-Priming ,Neoplasms ,Histocompatibility Antigens Class II ,Animals ,Female ,Dendritic Cells ,CD40 Antigens ,CD8-Positive T-Lymphocytes ,Signal Transduction - Abstract
Conventional type 1 dendritic cells (cDC1)
- Published
- 2019
13. Transcription factor Zeb2 regulates commitment to plasmacytoid dendritic cell and monocyte fate
- Author
-
Roxane Tussiwand, Wumesh Kc, Carlos G. Briseño, Yujiro Higashi, Arifumi Iwata, Malay Haldar, Gary E. Grajales-Reyes, Kenneth M. Murphy, Xiaodi Wu, Nicole M. Kretzer, and Theresa L. Murphy
- Subjects
Male ,0301 basic medicine ,Cytoplasm ,Neutrophils ,Context (language use) ,Plasmacytoid dendritic cell ,CD8-Positive T-Lymphocytes ,Biology ,Monocytes ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Bone Marrow ,medicine ,Animals ,Cell Lineage ,Progenitor cell ,Transcription factor ,Zinc Finger E-box Binding Homeobox 2 ,Progenitor ,Inflammation ,Cyclin-dependent kinase 1 ,Multidisciplinary ,Integrases ,Gene Expression Profiling ,Monocyte ,Dendritic Cells ,Biological Sciences ,Flow Cytometry ,Cell biology ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,Female ,Gene Deletion ,030215 immunology - Abstract
Dendritic cells (DCs) and monocytes develop from a series of bone-marrow–resident progenitors in which lineage potential is regulated by distinct transcription factors. Zeb2 is an E-box–binding protein associated with epithelial–mesenchymal transition and is widely expressed among hematopoietic lineages. Previously, we observed that Zeb2 expression is differentially regulated in progenitors committed to classical DC (cDC) subsets in vivo. Using systems for inducible gene deletion, we uncover a requirement for Zeb2 in the development of Ly-6Chi monocytes but not neutrophils, and we show a corresponding requirement for Zeb2 in expression of the M-CSF receptor in the bone marrow. In addition, we confirm a requirement for Zeb2 in development of plasmacytoid DCs but find that Zeb2 is not required for cDC2 development. Instead, Zeb2 may act to repress cDC1 progenitor specification in the context of inflammatory signals.
- Published
- 2016
- Full Text
- View/download PDF
14. RAB43 facilitates cross-presentation of cell-associated antigens by CD8α+ dendritic cells
- Author
-
Vivek Durai, Theresa L. Murphy, Roxane Tussiwand, Gary E. Grajales-Reyes, Kenneth M. Murphy, Nicole M. Kretzer, Xiaodi Wu, Prachi Bagadia, Carlos G. Briseño, Jörn C. Albring, and Derek J. Theisen
- Subjects
0301 basic medicine ,medicine.drug_class ,CD8 Antigens ,Immunology ,Cell ,Golgi Apparatus ,chemical and pharmacologic phenomena ,Biology ,Monoclonal antibody ,Monocytes ,Germline ,Mice ,03 medical and health sciences ,symbols.namesake ,Immune system ,Antigen ,medicine ,Animals ,Immunology and Allergy ,Research Articles ,Mice, Knockout ,Antigen Presentation ,Brief Definitive Report ,Cross-presentation ,hemic and immune systems ,Dendritic Cells ,Golgi apparatus ,Molecular biology ,In vitro ,3. Good health ,Repressor Proteins ,Basic-Leucine Zipper Transcription Factors ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,rab GTP-Binding Proteins ,symbols - Abstract
RAB43 is a vesicular transport protein unique to CD8α+ DCs that is localized to the Golgi. Kretzer et al. show that RAB43 is necessary for optimal cross-presentation of cell-associated antigens by CD8α+ DCs in vitro and in vivo but that it is dispensable for cross-presentation by in vitro monocyte-derived DCs., In this study, to examine cross-presentation by classical dendritic cells (DCs; cDCs), we evaluated the role of RAB43, a protein found to be selectively expressed by Batf3-dependent CD8α+ and CD103+ compared with other DC subsets and immune lineages. Using a specific monoclonal antibody, we localized RAB43 expression to the Golgi apparatus and LAMP1− cytoplasmic vesicles. Mice with germline or conditional deletion of Rab43 are viable and fertile and have normal development of cDCs but show a defect for in vivo and in vitro cross-presentation of cell-associated antigen. This defect is specific to cDCs, as Rab43-deficient monocyte-derived DCs showed no defect in cross-presentation of cell-associated antigen. These results suggest that RAB43 provides a specialized activity used in cross-presentation selectively by CD8α+ DCs but not other antigen-presenting cells.
- Published
- 2016
- Full Text
- View/download PDF
15. Transcriptional Control of Dendritic Cell Development
- Author
-
Vivek Durai, Theresa L. Murphy, Gary E. Grajales-Reyes, Arifumi Iwata, Carlos G. Briseño, Kenneth M. Murphy, Nicole M. Kretzer, Roxane Tussiwand, and Xiaodi Wu
- Subjects
Transcriptional Activation ,0301 basic medicine ,Myeloid ,Cellular differentiation ,Immunology ,Bone Marrow Cells ,chemical and pharmacologic phenomena ,Biology ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,medicine ,Transcriptional regulation ,Animals ,Humans ,Immunology and Allergy ,Cell Lineage ,Gene Regulatory Networks ,Regulation of gene expression ,Immunity, Cellular ,Effector ,Gene Expression Profiling ,Cell Differentiation ,Dendritic Cells ,Dendritic cell ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,Conventional Dendritic Cell ,030215 immunology - Abstract
The dendritic cells (DCs) of the immune system function in innate and adaptive responses by directing activity of various effector cells rather than serving as effectors themselves. DCs and closely related myeloid lineages share expression of many surface receptors, presenting a challenge in distinguishing their unique in vivo functions. Recent work has taken advantage of unique transcriptional programs to identify and manipulate murine DCs in vivo. This work has assigned several nonredundant in vivo functions to distinct DC lineages, consisting of plasmacytoid DCs and several subsets of classical DCs that promote different immune effector modules in response to pathogens. In parallel, a correspondence between human and murine DC subsets has emerged, underlying structural similarities for the DC lineages between these species. Recent work has begun to unravel the transcriptional circuitry that controls the development and diversification of DCs from common progenitors in the bone marrow.
- Published
- 2016
- Full Text
- View/download PDF
16. A Subset of Type I Conventional Dendritic Cells Controls Cutaneous Bacterial Infections through VEGF alpha-Mediated Recruitment of Neutrophils
- Author
-
Sandra Hubert, Martin Guilliams, Carlos G. Briseño, Benoit Malleret, Lai Guan Ng, Baptiste Janela, Mai Chan Lau, Josephine Lum, Kenneth M. Murphy, Philippe Musette, Yannick Simoni, Leen Vanhoutte, Bernard Malissen, Laurent F. Hennequin, Christian Wohn, Wan Ting Kong, Amit Patel, Michael G. Fehlings, Michael Poidinger, Rasha Msallam, Florent Ginhoux, Ansuman T. Satpathy, Emmanuel Vial, Jinmiao Chen, Chi Ching Goh, Francesca Zolezzi, Feriel Hacini-Rachinel, Evan W. Newell, Alexander A. Maini, Simon Yona, Agency for science, technology and research [Singapore] (A*STAR), Institut Necker Enfants-Malades (INEM - UM 111 (UMR 8253 / U1151)), Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), University of Toronto, Singapore Immunology Network (SIgN), Biomedical Sciences Institute (BMSI), Institut Cochin (IC UM3 (UMR 8104 / U1016)), Centre National de la Recherche Scientifique (CNRS)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), Department of Microbiology & Immunology [Singapore] (Yong Loo Lin School of Medicine), National University of Singapore (NUS), Department of Biotechnology and Biosciences, Università degli Studi di Milano-Bicocca [Milano] (UNIMIB), Washington University school of medicine, Centre d'Immunologie de Marseille - Luminy (CIML), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Aix Marseille Université (AMU), Universiteit Gent = Ghent University [Belgium] (UGENT), Virologie humaine, École normale supérieure - Lyon (ENS Lyon)-IFR128-Institut National de la Santé et de la Recherche Médicale (INSERM), Nestle Skin Health GALDERMA R&D, Service de Dermatologie [Rouen], CHU Rouen, Normandie Université (NU)-Normandie Université (NU)-Hôpital Charles Nicolle [Rouen], Galderma International S.A.S. [Paris, France], Università degli Studi di Milano-Bicocca = University of Milano-Bicocca (UNIMIB), Aix Marseille Université (AMU)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Universiteit Gent = Ghent University (UGENT), École normale supérieure de Lyon (ENS de Lyon)-IFR128-Institut National de la Santé et de la Recherche Médicale (INSERM), Hôpital Charles Nicolle [Rouen], Normandie Université (NU)-Normandie Université (NU)-CHU Rouen, Normandie Université (NU)-Normandie Université (NU), Genopolis Consortium, University of Milano-Bicocca, Ghent University [Belgium] (UGENT), and Hôpital Charles Nicolle [Rouen]-CHU Rouen
- Subjects
0301 basic medicine ,Vascular Endothelial Growth Factor A ,Injections, Intradermal ,Neutrophils ,medicine.medical_treatment ,Immunology ,Antigen presentation ,Biology ,03 medical and health sciences ,Propionibacterium acnes ,Mice ,0302 clinical medicine ,Immune system ,Immunity ,Acne Vulgaris ,medicine ,Immunology and Allergy ,Animals ,Humans ,RNA, Messenger ,Ear, External ,Gram-Positive Bacterial Infections ,Antigen Presentation ,integumentary system ,Monocyte ,Dendritic cell ,Dendritic Cells ,biology.organism_classification ,3. Good health ,Mice, Inbred C57BL ,Chemotaxis, Leukocyte ,030104 developmental biology ,Infectious Diseases ,medicine.anatomical_structure ,MRNA Sequencing ,Cytokine ,Gene Ontology ,Gene Expression Regulation ,Neutrophil Infiltration ,030220 oncology & carcinogenesis ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,Single-Cell Analysis - Abstract
Skin conventional dendritic cells (cDCs) exist as two distinct subsets, cDC1s and cDC2s, which maintain the balance of immunity to pathogens and tolerance to self and microbiota. Here, we examined the roles of dermal cDC1s and cDC2s during bacterial infection, notably Propionibacterium acnes (P. acnes). cDC1s, but not cDC2s, regulated the magnitude of the immune response to P. acnes in the murine dermis by controlling neutrophil recruitment to the inflamed site and survival and function therein. Single-cell mRNA sequencing revealed that this regulation relied on secretion of the cytokine vascular endothelial growth factor alpha (VEGF-alpha) by a minor subset of activated EpCAM(+)CD59(+) Ly-6D(+) cDC1s. Neutrophil recruitment by dermal cDC1s was also observed during S. aureus, bacillus Calmette-Guerin (BCG), or E. coli infection, as well as in a model of bacterial insult in human skin. Thus, skin cDC1s are essential regulators of the innate response in cutaneous immunity and have roles beyond classical antigen presentation.
- Published
- 2019
- Full Text
- View/download PDF
17. An Nfil3-Zeb2-Id2 pathway imposes Irf8 enhancer switching during cDC1 development
- Author
-
Theresa L. Murphy, Howard Y. Chang, Gary E. Grajales-Reyes, Zora Modrusan, Vivek Durai, Maximilian Nitschké, Jeffrey M. Granja, Xiao Huang, Carlos G. Briseño, Ansuman T. Satpathy, Marco Gargaro, Tiantian Liu, Kenneth M. Murphy, Arifumi Iwata, Sunkyung Kim, Andrey S. Shaw, and Prachi Bagadia
- Subjects
0301 basic medicine ,T cell ,Immunology ,Plasmacytoid dendritic cell ,Biology ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,medicine ,Immunology and Allergy ,Animals ,Progenitor cell ,Enhancer ,Transcription factor ,Cells, Cultured ,Inhibitor of Differentiation Protein 2 ,Zinc Finger E-box Binding Homeobox 2 ,Mice, Knockout ,Stem Cells ,NFIL3 ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Dendritic cell ,Dendritic Cells ,Cell biology ,Mice, Inbred C57BL ,Repressor Proteins ,030104 developmental biology ,medicine.anatomical_structure ,Basic-Leucine Zipper Transcription Factors ,Enhancer Elements, Genetic ,Interferon Regulatory Factors ,IRF8 ,030215 immunology - Abstract
Classical type 1 dendritic cells (cDC1s) are required for antiviral and antitumor immunity, which necessitates an understanding of their development. Development of the cDC1 progenitor requires an E-protein-dependent enhancer located 41 kilobases downstream of the transcription start site of the transcription factor Irf8 (+41-kb Irf8 enhancer), but its maturation instead requires the Batf3-dependent +32-kb Irf8 enhancer. To understand this switch, we performed single-cell RNA sequencing of the common dendritic cell progenitor (CDP) and identified a cluster of cells that expressed transcription factors that influence cDC1 development, such as Nfil3, Id2 and Zeb2. Genetic epistasis among these factors revealed that Nfil3 expression is required for the transition from Zeb2hi and Id2lo CDPs to Zeb2lo and Id2hi CDPs, which represent the earliest committed cDC1 progenitors. This genetic circuit blocks E-protein activity to exclude plasmacytoid dendritic cell potential and explains the switch in Irf8 enhancer usage during cDC1 development. Classical type 1 dendritic cells (cDC1s) are essential for activation of antiviral and anticancer T cell responses. Murphy and colleagues describe a genetic circuit that involves the transcription factors Nfil3, Id2 and Zeb2. This circuit imposes a molecular switch that allows cDC1 specification and development.
- Published
- 2018
18. Altered compensatory cytokine signaling underlies the discrepancy between Flt3-/- and Flt3l-/- mice
- Author
-
Daved H. Fremont, Vivek Durai, Jesse T. Davidson, Kenneth M. Murphy, Derek J. Theisen, Prachi Bagadia, Carlos G. Briseño, Marco Gargaro, Theresa L. Murphy, and Arifumi Iwata
- Subjects
0301 basic medicine ,Macrophage colony-stimulating factor ,Immunology and Allergy ,Immunology ,Chemistry ,medicine.medical_treatment ,hemic and immune systems ,Stem cell factor ,Dendritic cell ,Cell biology ,03 medical and health sciences ,fluids and secretions ,030104 developmental biology ,Cytokine ,hemic and lymphatic diseases ,embryonic structures ,Fms-Like Tyrosine Kinase 3 ,medicine ,Progenitor cell ,Signal transduction ,Receptor - Abstract
The receptor Flt3 and its ligand Flt3L are both critical for dendritic cell (DC) development, but DC deficiency is more severe in Flt3l−/− mice than in Flt3−/− mice. This has led to speculation that Flt3L binds to another receptor that also supports DC development. However, we found that Flt3L administration does not generate DCs in Flt3−/− mice, arguing against a second receptor. Instead, Flt3−/− DC progenitors matured in response to macrophage colony–stimulating factor (M-CSF) or stem cell factor, and deletion of Csf1r in Flt3−/− mice further reduced DC development, indicating that these cytokines could compensate for Flt3. Surprisingly, Flt3−/− DC progenitors displayed enhanced M-CSF signaling, suggesting that loss of Flt3 increased responsiveness to other cytokines. In agreement, deletion of Flt3 in Flt3l−/− mice paradoxically rescued their severe DC deficiency. Thus, multiple cytokines can support DC development, and the discrepancy between Flt3−/− and Flt3l−/− mice results from the increased sensitivity of Flt3−/− progenitors to these cytokines.
- Published
- 2018
19. Development, Diversity, and Function of Dendritic Cells in Mouse and Human
- Author
-
Carlos G. Briseño, David A. Anderson, and Kenneth M. Murphy
- Subjects
0301 basic medicine ,Lineage (genetic) ,Cellular differentiation ,medicine.medical_treatment ,Bone Marrow Cells ,Biology ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,Antigen ,medicine ,Animals ,Humans ,Transcription factor ,Cell Differentiation ,Dendritic cell ,Dendritic Cells ,Cell biology ,030104 developmental biology ,Cytokine ,Gene Expression Regulation ,PERSPECTIVES ,Cytokines ,Function (biology) ,030215 immunology - Abstract
The study of murine dendritic cell (DC) development has been integral to the identification of specialized DC subsets that have unique requirements for their form and function. Advances in the field have also provided a framework for the identification of human DC counterparts, which appear to have conserved mechanisms of development and function. Multiple transcription factors are expressed in unique combinations that direct the development of classical DCs (cDCs), which include two major subsets known as cDC1s and cDC2s, and plasmacytoid DCs (pDCs). pDCs are potent producers of type I interferons and thus these cells are implicated in immune responses that depend on this cytokine. Mouse models deficient in the cDC1 lineage have revealed their importance in directing immune responses to intracellular bacteria, viruses, and cancer through the cross-presentation of cell-associated antigen. Models of transcription factor deficiency have been used to identify subsets of cDC2 that are required for T helper (Th)2 and Th17 responses to certain pathogens; however, no single factor is known to be absolutely required for the development of the complete cDC2 lineage. In this review, we will discuss the current state of knowledge of mouse and human DC development and function and highlight areas in the field that remain unresolved.
- Published
- 2017
20. Altered compensatory cytokine signaling underlies the discrepancy between
- Author
-
Vivek, Durai, Prachi, Bagadia, Carlos G, Briseño, Derek J, Theisen, Arifumi, Iwata, Jesse T, Davidson, Marco, Gargaro, Daved H, Fremont, Theresa L, Murphy, and Kenneth M, Murphy
- Subjects
Stem Cell Factor ,Transcription, Genetic ,Stem Cells ,Granulocyte-Macrophage Colony-Stimulating Factor ,Membrane Proteins ,hemic and immune systems ,Receptor, Macrophage Colony-Stimulating Factor ,Dendritic Cells ,Article ,Mice, Inbred C57BL ,Proto-Oncogene Proteins c-kit ,fluids and secretions ,fms-Like Tyrosine Kinase 3 ,Bone Marrow ,hemic and lymphatic diseases ,embryonic structures ,Animals ,Cytokines ,Gene Deletion ,Research Articles ,Signal Transduction - Abstract
Durai et al. demonstrate that the less severe DC deficiency in Flt3–/– mice compared to Flt3l–/– mice is due to the enhanced sensitivity of Flt3–/– progenitors to compensatory cytokines that support DC development., The receptor Flt3 and its ligand Flt3L are both critical for dendritic cell (DC) development, but DC deficiency is more severe in Flt3l−/− mice than in Flt3−/− mice. This has led to speculation that Flt3L binds to another receptor that also supports DC development. However, we found that Flt3L administration does not generate DCs in Flt3−/− mice, arguing against a second receptor. Instead, Flt3−/− DC progenitors matured in response to macrophage colony–stimulating factor (M-CSF) or stem cell factor, and deletion of Csf1r in Flt3−/− mice further reduced DC development, indicating that these cytokines could compensate for Flt3. Surprisingly, Flt3−/− DC progenitors displayed enhanced M-CSF signaling, suggesting that loss of Flt3 increased responsiveness to other cytokines. In agreement, deletion of Flt3 in Flt3l−/− mice paradoxically rescued their severe DC deficiency. Thus, multiple cytokines can support DC development, and the discrepancy between Flt3−/− and Flt3l−/− mice results from the increased sensitivity of Flt3−/− progenitors to these cytokines.
- Published
- 2017
21. Deficiency of transcription factor RelB perturbs myeloid and DC development by hematopoietic-extrinsic mechanisms
- Author
-
David A. Anderson, Vivek Durai, Carlos G. Briseño, Jesse T. Davidson, Theresa L. Murphy, Deborah V. Novack, Kenneth M. Murphy, Derek J. Theisen, and Marco Gargaro
- Subjects
0301 basic medicine ,CD4-Positive T-Lymphocytes ,Lymphotoxin-beta ,Myeloid ,Hematopoietic System ,Transcription Factor RelB ,CD8-Positive T-Lymphocytes ,Protein Serine-Threonine Kinases ,03 medical and health sciences ,0302 clinical medicine ,Lymphotoxin beta Receptor ,transcription factors ,medicine ,Animals ,Myeloid Cells ,Transcription factor ,Multidisciplinary ,CD40 ,biology ,RELB ,Dendritic Cells ,Biological Sciences ,dendritic cells ,hematopoiesis ,hematopoietic niche ,Mice, Mutant Strains ,Cell biology ,Mice, Inbred C57BL ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,Lymphotoxin ,biology.protein ,Myelopoiesis ,Spleen ,030215 immunology - Abstract
RelB is an NF-κB family transcription factor activated in the noncanonical pathway downstream of NF-κB-inducing kinase (NIK) and TNF receptor family members including lymphotoxin-β receptor (LTβR) and CD40. Early analysis suggested that RelB is required for classical dendritic cell (cDC) development based on a severe reduction of cDCs in Relb-/- mice associated with profound myeloid expansion and perturbations in B and T cells. Subsequent analysis of radiation chimeras generated from wild-type and Relb-/- bone marrow showed that RelB exerts cell-extrinsic actions on some lineages, but it has remained unclear whether the impact of RelB on cDC development is cell-intrinsic or -extrinsic. Here, we reevaluated the role of RelB in cDC and myeloid development using a series of radiation chimeras. We found that there was no cell-intrinsic requirement for RelB for development of most cDC subsets, except for the Notch2- and LTβR-dependent subset of splenic CD4+ cDC2s. These results identify a relatively restricted role of RelB in DC development. Moreover, the myeloid expansion in Relb-/- mice resulted from hematopoietic-extrinsic actions of RelB. This result suggests that there is an unrecognized but critical role for RelB within the nonhematopoietic niche that controls normal myelopoiesis.
- Published
- 2017
22. Wdfy4-deficiency reveals a critical role for cross-presentation in anti-viral and anti-tumor responses
- Author
-
Derek J Theisen, Jesse T Davidson, Carlos G Briseño, Marco Gargaro, Elvin J Lauron, Qiuling Wang, Pritesh Desai, Vivek Durai, Prachi Bagadia, Joshua R Brickner, Wandy L Beatty, Herbert W Virgin, William E Gillanders, Nima Mossamaparast, Michael S Diamond, L David Sibley, Wayne Yokoyama, Robert D Schreiber, Theresa L Murphy, and Kenneth M Murphy
- Subjects
Immunology ,Immunology and Allergy - Abstract
Cross-presentation allows for the presentation of exogenous viral and tumor antigens to CD8+ T cells by XCR1+ classical dendritic cells (cDC1s). Previously, the role of cross-presentation in cytotoxic T cell responses in vivo was unclear due to the lack of mouse models specifically deficient in cross-presentation. We developed a CRISPR-Cas9 screen to find novel regulators of cross-presentation and found that the protein WDFY4 is essential for cross-presentation of cell-associated antigens but not for direct presentation or presentation on MHCII. WDFY4-deficient mice had normal cDC1 development and were able to survive infections with Toxoplasma gondii through production of IL-12. However, WDFY4-deficient mice failed to prime CD8+ T cells to viral antigens and were completely unable to control immunogenic tumors. WDFY4 interacts with proteins related to vesicular formation and trafficking such as clathrin and HSP90ab1 and localizes near the cell surface, suggesting its involvement in vesicular targeting after antigen uptake during cross-presentation.
- Published
- 2019
- Full Text
- View/download PDF
23. Notch2-dependent classical dendritic cells orchestrate intestinal immunity against attaching and effacing bacterial pathogens
- Author
-
Wan Ling Lee, Carlos G. Briseño, Raphael Kopan, Jennifer L. Gommerman, Mustafa Turkoz, Nicholas A. Manieri, Dennis K. P. Ng, Marco Colonna, Theresa L. Murphy, Stephanie R. Thomas, Wumesh Kc, Jacob S. Lee, Michel C. Nussenzweig, Kenneth M. Murphy, Xiaodi Wu, Christina Song, Cynthia J. Guidos, Rodney D. Newberry, Keely G. McDonald, Ansuman T. Satpathy, Wenjun Ouyang, Matthew M. Meredith, and Thaddeus S. Stappenbeck
- Subjects
Cellular differentiation ,Immunology ,Mice, Transgenic ,Receptors, Cell Surface ,Biology ,Interleukin-23 ,Article ,Microbiology ,Minor Histocompatibility Antigens ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Intestinal mucosa ,Immunity ,Antigens, CD ,Lymphotoxin beta Receptor ,Citrobacter rodentium ,Immunology and Allergy ,Animals ,Lectins, C-Type ,Receptor, Notch2 ,Intestinal Mucosa ,030304 developmental biology ,0303 health sciences ,Wound Healing ,Innate immune system ,CD11b Antigen ,Innate lymphoid cell ,Enterobacteriaceae Infections ,Cell Differentiation ,Dendritic Cells ,3. Good health ,Mucosal immunology ,Host-Pathogen Interactions ,Lymphotoxin beta receptor ,Spleen ,030215 immunology ,Signal Transduction ,Transcription Factors - Abstract
Defense against attaching-and-effacing bacteria requires the sequential generation of interleukin 23 (IL-23) and IL-22 to induce protective mucosal responses. Although CD4(+) and NKp46(+) innate lymphoid cells (ILCs) are the critical source of IL-22 during infection, the precise source of IL-23 is unclear. We used genetic techniques to deplete mice of specific subsets of classical dendritic cells (cDCs) and analyzed immunity to the attaching-and-effacing pathogen Citrobacter rodentium. We found that the signaling receptor Notch2 controlled the terminal stage of cDC differentiation. Notch2-dependent intestinal CD11b(+) cDCs were an obligate source of IL-23 required for survival after infection with C. rodentium, but CD103(+) cDCs dependent on the transcription factor Batf3 were not. Our results demonstrate a nonredundant function for CD11b(+) cDCs in the response to pathogens in vivo.
- Published
- 2013
24. CAR-T cells and CAR-Tregs targeting conventional type-1 dendritic cell suppress experimental autoimmune encephalomyelitis
- Author
-
Cody D. Moorman, Sherman Yu, Carlos G. Briseno, Hyewon Phee, Anupama Sahoo, Ambika Ramrakhiani, and Ashutosh Chaudhry
- Subjects
autoimmune disease ,autoimmunity ,chimeric antigen receptor ,CAR-T cells ,CAR-Treg ,type 1 conventional dendritic cells ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Conventional type 1 dendritic cells (DC1) contribute to the development of pathogenic T helper type 1 (Th1) cells in part via the production of the proinflammatory cytokine interleukin-12. Thus, depletion of DC1 has the potential to dampen autoimmune responses. Here, we developed X-C motif chemokine receptor 1 (XCR1)-specific chimeric antigen receptor (CAR)-T cells and CAR-Tregs that specifically targeted DC1. XCR1 CAR-T cells were successfully generated as CD4+ and CD8+ T cells, expressed XCR1 CAR efficiently, and induced XCR1-dependent activation, cytokine production and proliferation. XCR1 CAR-T cells selectively depleted DC1 when transferred into RAG2−/− mice with a compensatory increase in conventional type 2 DC (DC2) and plasmacytoid DC (pDC). XCR1 CAR-T cell-mediated depletion of DC1 modestly suppressed the onset of Th1-driven experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. Diphtheria toxin-mediated DC1 depletion in XCR1-diphtheria toxin receptor mice also suppressed EAE, suggesting that DC1 depletion was responsible for EAE suppression. XCR1 CAR-Tregs were successfully generated and suppressed effector T cells in the presence of XCR1+ cells. Therapeutic treatment with XCR1 CAR-Tregs suppressed Th1-driven EAE. Therefore, we conclude that depletion of DC1 with XCR1 CAR-T cells or immune suppression with XCR1 CAR-Tregs can modestly suppress Th1-driven EAE.
- Published
- 2023
- Full Text
- View/download PDF
25. Quality of TCR signaling determined by differential affinities of enhancers for the composite BATF-IRF4 transcription factor complex
- Author
-
Vivek Durai, Theresa L. Murphy, Kenneth M. Murphy, Gary E. Grajales-Reyes, Carlos G. Briseño, Arifumi Iwata, Takeshi Egawa, Xiaodi Wu, and Roxane Tussiwand
- Subjects
0301 basic medicine ,Mice, 129 Strain ,Receptors, Antigen, T-Cell, alpha-beta ,Immunology ,Receptors, Antigen, T-Cell ,Transcription factor complex ,chemical and pharmacologic phenomena ,Stimulation ,Autoimmunity ,Biology ,Polymorphism, Single Nucleotide ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Th2 Cells ,BATF ,Immunology and Allergy ,Animals ,Humans ,CTLA-4 Antigen ,Genetic Predisposition to Disease ,Enhancer ,Gene ,Genetics ,Mice, Knockout ,Mice, Inbred BALB C ,T-cell receptor ,Cell Differentiation ,Affinities ,Cell biology ,030104 developmental biology ,Basic-Leucine Zipper Transcription Factors ,Enhancer Elements, Genetic ,Multiprotein Complexes ,Interferon Regulatory Factors ,030215 immunology ,IRF4 ,Protein Binding ,Signal Transduction - Abstract
Variable strengths of T cell receptor (TCR) signaling can produce divergent outcomes, but the mechanism remains obscure. The abundance of the transcription factor IRF4 increases with TCR signal strength, but how this would induce distinct types of responses is unclear. We compared TH2 gene expression with BATF/IRF4 enhancer occupancy at varying strengths of TCR stimulation. BATF/IRF4-dependent genes clustered into distinct TCR-sensitivities. Enhancers exhibited a spectrum of occupancy by BATF/IRF4 ternary complex that correlated with TCR-sensitivity of gene expression. DNA sequences immediately flanking the previously defined AICE motif controlled the affinity for BATF/IRF4 for direct binding to DNA. ChIP-exo analysis allowed identification of a novel high-affinity AICE2 motif at a human SNP of CTLA4 associated with resistance to autoimmunity. Thus, the affinity of different enhancers for the BATF-IRF4 complex may underlie divergent signaling outcomes in response to various strengths of TCR signaling.
- Published
- 2016
26. Mafb lineage tracing to distinguish macrophages from other immune lineages reveals dual identity of Langerhans cells
- Author
-
Jörn C. Albring, Carlos G. Briseño, Vivek Durai, Theresa L. Murphy, Gwendalyn J. Randolph, Malay Haldar, Prachi Bagadia, Ki-Wook Kim, Kenneth M. Murphy, and Xiaodi Wu
- Subjects
0301 basic medicine ,Lineage (genetic) ,Immunology ,Population ,MafB Transcription Factor ,Biology ,Models, Biological ,Monocytes ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Antigen ,Immunology and Allergy ,Animals ,Antigens, Ly ,Cell Lineage ,Gene Knock-In Techniques ,education ,Lung ,Research Articles ,education.field_of_study ,Integrases ,Macrophages ,Brief Definitive Report ,Pneumonia ,Phenotype ,3. Good health ,Cell biology ,Hematopoiesis ,Mice, Inbred C57BL ,Haematopoiesis ,030104 developmental biology ,MAFB ,Organ Specificity ,Langerhans Cells ,Gene Targeting ,030215 immunology ,Transcription Factors - Abstract
Using Mafb-driven Cre, Murphy et al. establish a new tool to discriminate macrophages from other myeloid cells in vivo., Current systems for conditional gene deletion within mouse macrophage lineages are limited by ectopic activity or low efficiency. In this study, we generated a Mafb-driven Cre strain to determine whether any dendritic cells (DCs) identified by Zbtb46-GFP expression originate from a Mafb-expressing population. Lineage tracing distinguished macrophages from classical DCs, neutrophils, and B cells in all organs examined. At steady state, Langerhans cells (LCs) were lineage traced but also expressed Zbtb46-GFP, a phenotype not observed in any other population. After exposure to house dust mite antigen, Zbtb46-negative CD64+ inflammatory cells infiltrating the lung were substantially lineage traced, but Zbtb46-positive CD64− cells were not. These results provide new evidence for the unique identity of LCs and challenge the notion that some inflammatory cells are a population of monocyte-derived DCs.
- Published
- 2016
27. CCR7 and IRF4-dependent dendritic cells regulate lymphatic collecting vessel permeability
- Author
-
Stoyan Ivanov, Peter L. Wang, Ki-Wook Kim, Brian Saunders, Michael J. Davis, Jesse W. Williams, Emma L. Kuan, Brant E. Isakson, Gwendalyn J. Randolph, Bernd H. Zinselmeyer, Erica G. Weinstein, Carlos G. Briseño, Melissa Ouhachi, Reinhold Förster, Adam C. Straub, Joshua P. Scallan, Emmanuel L. Gautier, Michael W. Johnson, Marco Colonna, Kathrin Werth, Washington University School of Medicine in St. Louis, Washington University in Saint Louis (WUSTL), University of Missouri [Columbia] (Mizzou), University of Missouri System, Hannover Medical School [Hannover] (MHH), Icahn School of Medicine at Mount Sinai [New York] (MSSM), University of Virginia School of Medicine [US], Unité de Recherche sur les Maladies Cardiovasculaires, du Métabolisme et de la Nutrition = Research Unit on Cardiovascular and Metabolic Diseases (ICAN), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Institut de Cardiométabolisme et Nutrition = Institute of Cardiometabolism and Nutrition [CHU Pitié Salpêtrière] (IHU ICAN), CHU Pitié-Salpêtrière [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-CHU Pitié-Salpêtrière [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU), Gautier, Emmanuel, Washington University school of medicine, University of Virginia School of Medicine, Unité de Recherche sur les Maladies Cardiovasculaires, du Métabolisme et de la Nutrition = Institute of cardiometabolism and nutrition (ICAN), Université Pierre et Marie Curie - Paris 6 (UPMC)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-CHU Pitié-Salpêtrière [AP-HP], Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU), HAL UPMC, Gestionnaire, and Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Sorbonne Université (SU)
- Subjects
0301 basic medicine ,Receptors, CCR7 ,Pathology ,medicine.medical_specialty ,[SDV.IMM] Life Sciences [q-bio]/Immunology ,MESH: Lymphatic Vessels ,[SDV]Life Sciences [q-bio] ,Antigen presentation ,chemical and pharmacologic phenomena ,C-C chemokine receptor type 7 ,Vascular permeability ,MESH: Mice, Knockout ,MESH: Receptors, CCR7 ,Capillary Permeability ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Fibrosis ,medicine ,Animals ,MESH: Animals ,MESH: Mice ,Lymphatic Vessels ,Mice, Knockout ,MESH: Dendritic Cells ,business.industry ,MESH: Capillary Permeability ,hemic and immune systems ,Dendritic Cells ,General Medicine ,medicine.disease ,3. Good health ,[SDV] Life Sciences [q-bio] ,030104 developmental biology ,Lymphatic system ,Interferon Regulatory Factors ,MESH: Fibrosis ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,Lymph ,business ,MESH: Interferon Regulatory Factors ,Research Article ,030215 immunology ,IRF4 - Abstract
International audience; Lymphatic collecting vessels direct lymph into and from lymph nodes (LNs) and can become hyperpermeable as the result of a previous infection. Enhanced permeability has been implicated in compromised immunity due to reduced flow of lymph and immune cells to LNs, which are the primary site of antigen presentation to T cells. Presently, very little is known about the molecular signals that affect lymphatic collecting vessel permeability. Here, we have shown that lymphatic collecting vessel permeability is controlled by CCR7 and that the chronic hyperpermeability of collecting vessels observed in Ccr7–/– mice is followed by vessel fibrosis. Reexpression of CCR7 in DCs, however, was sufficient to reverse the development of such fibrosis. IFN regulatory factor 4–positive (IRF4+) DCs constitutively interacted with collecting lymphatics, and selective ablation of this DC subset in Cd11c-Cre Irf4fl/fl mice also rendered lymphatic collecting vessels hyperpermeable and fibrotic. Together, our data reveal that CCR7 plays multifaceted roles in regulating collecting vessel permeability and fibrosis, with one of the key players being IRF4-dependent DCs.
- Published
- 2016
- Full Text
- View/download PDF
28. Abstract 3781: A focused dendritic cell CRISPR/Cas9 screen to identify novel regulators of cross-presentation and antitumor immunity
- Author
-
Vivek Durai, Derek J. Theisen, Kenneth M. Murphy, Carlos G. Briseño, William E. Gillanders, Jesse T. Davidson, and Theresa L. Murphy
- Subjects
Cancer Research ,Immune system ,Oncology ,Antigen ,Cancer research ,Cytotoxic T cell ,Cross-presentation ,Dendritic cell ,Biology ,Acquired immune system ,CD8 ,Viral vector - Abstract
Introduction: Dendritic cells (DCs) are professional antigen-presenting cells with two major subsets, the classical DC type 1 and 2 (cDC1 and cDC2). The cDC1, also known as the CD8α+ DC, is especially equipped for the process of cross-presentation in which dead cell antigen is presented on MHC-I to prime cytotoxic T cells. This process is relevant to anti-cancer immunity, as mice deficient in the transcription factor BATF3, which specifically lack the cDC1, succumb to highly immunogenic tumors that are normally rejected and cleared in wild-type mice. The mechanism of cross-presentation by the cDC1 is poorly understood. In this study, we developed a focused CRISPR/Cas9 screen to identify novel genes that endow the cDC1 with the ability to cross-prime cytotoxic T cells. Methods: A curated list of target cDC1 genes was constructed using the Immunological Genome Project Database as the primary resource (www.immgen.org). 120 genes specifically expressed by the cDC1 in comparison to other murine immune cells were selected. Guide RNAs (gRNAs) were designed to early exon sequences using the MIT CRISPR Design Tool to minimize off-target effects (www.crispr.mit.edu), and 2-3 different gRNAs were evaluated in tandem for each candidate. These gRNAs were transduced via retroviral vector into lineage depleted c-Kithi bone marrow cells isolated from C57BL/6J Cas9 knock-in mice. These cells were differentiated in vitro for 8 days in the presence of the cytokine Flt3L. Transduced DCs were sorted by FACS and co-cultured in the presence of a source of dead cell antigen (heat-killed L. monocytogenes expressing ovalbumin, or HKLM-OVA) as well as CFSE-labelled transgenic CD8+ T cells with an ovalbumin peptide-specific T cell receptor (OT-1). The proliferation of OT-1 cells was measured by CFSE dilution. Results: To validate our CRISPR/Cas9-based screening approach, we targeted the first exon of the beta-2 microglobulin gene (B2M), which is required for cell surface expression of MHC-I. B2M knockout DCs were co-cultured in the presence of HKLM-OVA and CSFE-labelled OT-1 cells for 3 days. Controls included DCs transduced with a scramble gRNA with or without antigen. As expected, targeting B2M in DCs led to near-complete reduction in OT-1 proliferation compared to scramble control (3.7% vs. 20.6%, p=0.003), similar to the absence of antigen (3.7% vs. 0.96%, p=0.99). We then expanded our assay to our curated list. Promising candidate genes have been discovered and are currently being validated in murine knockout models. Conclusions: The CRISPR/Cas9 system can be adapted to screen the cDC1 for novel genes involved in cross-priming cytotoxic T cells. Validation of candidates in vivo, including tumor assays, will be presented at the meeting. These findings may have important implications for adaptive immunity against cancer. Citation Format: Jesse T. Davidson, Derek J. Theisen, Carlos G. Briseño, Vivek Durai, William E. Gillanders, Theresa L. Murphy, Kenneth M. Murphy. A focused dendritic cell CRISPR/Cas9 screen to identify novel regulators of cross-presentation and antitumor immunity [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 3781.
- Published
- 2018
- Full Text
- View/download PDF
29. Oral Antibiotic Treatment of Mice Exacerbates the Disease Severity of Multiple Flavivirus Infections
- Author
-
Herbert W. Virgin, Kenneth M. Murphy, Scott A. Handley, Barry L. Hykes, Jeffrey I. Gordon, Qing Tan, Tiffany M. Lucas, Derek J. Theisen, Chandni Desai, Michael S. Diamond, Prachi Bagadia, Hueylie Lin, Matthew J. Gorman, Subhajit Poddar, Carlos G. Briseño, and Larissa B. Thackray
- Subjects
0301 basic medicine ,Male ,medicine.drug_class ,viruses ,T-Lymphocytes ,Antibiotics ,Administration, Oral ,Gut flora ,Dengue virus ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,Article ,Dengue fever ,03 medical and health sciences ,Mice ,Immunity ,Aedes ,Immunopathology ,Chlorocebus aethiops ,medicine ,Animals ,Flavivirus Infections ,lcsh:QH301-705.5 ,Cecum ,Vero Cells ,biology ,business.industry ,Zika Virus Infection ,Flavivirus ,Microbiota ,virus diseases ,biology.organism_classification ,medicine.disease ,3. Good health ,Anti-Bacterial Agents ,Mice, Inbred C57BL ,030104 developmental biology ,lcsh:Biology (General) ,Immunology ,Ampicillin ,Female ,business - Abstract
SUMMARY Although the outcome of flavivirus infection can vary from asymptomatic to lethal, environmental factors modulating disease severity are poorly defined. Here, we observed increased susceptibility of mice to severe West Nile (WNV), Dengue, and Zika virus infections after treatment with oral antibiotics (Abx) that depleted the gut microbiota. Abx treatment impaired the development of optimal T cell responses, with decreased levels of WNV-specific CD8+ T cells associated with increased infection and immunopathology. Abx treatments that resulted in enhanced WNV susceptibility generated changes in the overall structure of the gut bacterial community and in the abundance of specific bacterial taxa. As little as 3 days of treatment with ampicillin was sufficient to alter host immunity and WNV outcome. Our results identify oral Abx therapy as a potential environmental determinant of systemic viral disease, and they raise the possibility that perturbation of the gut microbiota may have deleterious consequences for subsequent flavivirus infections., In Brief Thackray et al. observed increased susceptibility to West Nile, Zika, and Dengue virus infections following oral antibiotic treatment in mice. Antibiotics altered the bacterial abundance and community structure and the development of optimal T cell immunity. These data suggest that antibiotics may have deleterious consequences for subsequent flavivirus infections.
- Published
- 2018
30. Correction: Corrigendum: Regulation of monocyte cell fate by blood vessels mediated by Notch signalling
- Author
-
Jasmin Jussofie, Hermann Haller, L. Christian Napp, Christian Kupatt, Chieko Ishifune, Kashyap Krishnasamy, Johann Bauersachs, Freddy Radtke, Ursula Zimber-Strobl, Christian Weber, Florian P. Limbourg, Ralf H. Adams, Roberto Giagnorio, Oliver Soehnlein, Jaba Gamrekelashvili, Koji Yasutomo, Christine Häger, Rabea Hinkel, Carlos G. Briseño, Kenneth M. Murphy, Anne Limbourg, Saravana K. Ramasamy, Lothar J. Strobl, Tamar Kapanadze, and Johan Duchene
- Subjects
0301 basic medicine ,Multidisciplinary ,Monocyte ,Published Erratum ,Science ,Notch signaling pathway ,General Physics and Astronomy ,General Chemistry ,Biology ,Cell fate determination ,General Biochemistry, Genetics and Molecular Biology ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,medicine - Abstract
Nature Communications 7: Article number: 12597 (2016); Published: 31 August 2016; Updated: 3 May 2017 The authors inadvertently omitted Christine Häger, who was involved in the initial characterization of Notch mutant mice presented in this Article, from the author list and Author contributions statement.
- Published
- 2017
31. Batf3 maintains autoactivation of Irf8 for commitment of a CD8α(+) conventional DC clonogenic progenitor
- Author
-
Vivek Durai, Kenneth M. Murphy, Xiaodi Wu, Nicole M. Kretzer, Jörg Schönheit, Theresa L. Murphy, Gary E. Grajales-Reyes, Roxane Tussiwand, Wumesh Kc, Carlos G. Briseño, Jörn C. Albring, Frank Rosenbauer, Prachi Bagadia, Malay Haldar, and Arifumi Iwata
- Subjects
Mice, 129 Strain ,CD8 Antigens ,Immunology ,Molecular Sequence Data ,Bone Marrow Cells ,Mice, Transgenic ,Biology ,Sequence Homology, Nucleic Acid ,Immunology and Allergy ,Animals ,Receptors, Immunologic ,Clonogenic assay ,Transcription factor ,Cells, Cultured ,Progenitor ,Oligonucleotide Array Sequence Analysis ,Mice, Knockout ,Base Sequence ,Stem Cells ,CD24 Antigen ,Dendritic Cells ,Flow Cytometry ,Clone Cells ,Mice, Inbred C57BL ,Repressor Proteins ,Basic-Leucine Zipper Transcription Factors ,Interferon Regulatory Factors ,Cancer research ,Myelopoiesis ,IRF8 ,Transcriptome ,Conventional Dendritic Cell ,Protein Binding - Abstract
The transcription factors Batf3 and IRF8 are required for the development of CD8α(+) conventional dendritic cells (cDCs), but the basis for their actions has remained unclear. Here we identified two progenitor cells positive for the transcription factor Zbtb46 that separately generated CD8α(+) cDCs and CD4(+) cDCs and arose directly from the common DC progenitor (CDP). Irf8 expression in CDPs required prior autoactivation of Irf8 that was dependent on the transcription factor PU.1. Specification of the clonogenic progenitor of CD8α(+) cDCs (the pre-CD8 DC) required IRF8 but not Batf3. However, after specification of pre-CD8 DCs, autoactivation of Irf8 became Batf3 dependent at a CD8α(+) cDC-specific enhancer with multiple transcription factor AP1-IRF composite elements (AICEs) within the Irf8 superenhancer. CDPs from Batf3(-/-) mice that were specified toward development into pre-CD8 DCs failed to complete their development into CD8α(+) cDCs due to decay of Irf8 autoactivation and diverted to the CD4(+) cDC lineage.
- Published
- 2014
32. Runx1 and Cbfβ regulate the development of Flt3+ dendritic cell progenitors and restrict myeloproliferative disorder
- Author
-
Carlos G. Briseño, Chun Chou, Takeshi Egawa, Xiongwei Cai, Drew G. Michael, Ansuman T. Satpathy, Nancy A. Speck, Sunnie Hsiung, and Deepta Bhattacharya
- Subjects
Hematopoiesis and Stem Cells ,Cellular differentiation ,Immunology ,Gene Expression ,Bone Marrow Cells ,Core Binding Factor Alpha 1 Subunit ,Mice, Transgenic ,Biology ,Biochemistry ,Core Binding Factor beta Subunit ,chemistry.chemical_compound ,Mice ,Myeloproliferative Disorders ,medicine ,Animals ,Progenitor cell ,Cells, Cultured ,Mice, Knockout ,Reverse Transcriptase Polymerase Chain Reaction ,Stem Cells ,Anemia ,Cell Differentiation ,Cell Biology ,Hematology ,Dendritic Cells ,Flow Cytometry ,Cell biology ,Hematopoiesis ,Mice, Inbred C57BL ,Haematopoiesis ,medicine.anatomical_structure ,Core Binding Factor Alpha 3 Subunit ,RUNX1 ,chemistry ,fms-Like Tyrosine Kinase 3 ,Fms-Like Tyrosine Kinase 3 ,Core Binding Factor Alpha 2 Subunit ,Interferon Regulatory Factors ,Bone marrow ,IRF8 ,Granulocytes - Abstract
Runx1 and Cbfβ are critical for the establishment of definitive hematopoiesis and are implicated in leukemic transformation. Despite the absolute requirements for these factors in the development of hematopoietic stem cells and lymphocytes, their roles in the development of bone marrow progenitor subsets have not been defined. Here, we demonstrate that Cbfβ is essential for the development of Flt3(+) macrophage-dendritic cell (DC) progenitors in the bone marrow and all DC subsets in the periphery. Besides the loss of DC progenitors, pan-hematopoietic Cbfb-deficient mice also lack CD105(+) erythroid progenitors, leading to severe anemia at 3 to 4 months of age. Instead, Cbfb deficiency results in aberrant progenitor differentiation toward granulocyte-macrophage progenitors (GMPs), resulting in a myeloproliferative phenotype with accumulation of GMPs in the periphery and cellular infiltration of the liver. Expression of the transcription factor Irf8 is severely reduced in Cbfb-deficient progenitors, and overexpression of Irf8 restors DC differentiation. These results demonstrate that Runx proteins and Cbfβ restrict granulocyte lineage commitment to facilitate multilineage hematopoietic differentiation and thus identify their novel tumor suppressor function in myeloid leukemia.
- Published
- 2014
33. Complementary diversification of dendritic cells and innate lymphoid cells
- Author
-
Theresa L. Murphy, Carlos G. Briseño, and Kenneth M. Murphy
- Subjects
medicine.medical_treatment ,T cell ,Immunology ,chemical and pharmacologic phenomena ,Biology ,Article ,Immune system ,Immunity ,Bone Marrow ,medicine ,Immunology and Allergy ,Animals ,Homeostasis ,Humans ,Lymphocytes ,Antigen-presenting cell ,Innate immune system ,Innate lymphoid cell ,CCL18 ,Dendritic Cells ,Immunity, Innate ,Cell biology ,Cytokine ,medicine.anatomical_structure ,Cytokines - Abstract
Dendritic cells (DCs) are professional antigen presenting cells conventionally thought to mediate cellular adaptive immune responses. Recent studies have led to the recognition of a non-redundant role for DCs in orchestrating innate immune responses, and in particular, for DC subset-specific interactions with innate lymphoid cells (ILCs). Recently recognized as important effectors of early immune responses, ILCs develop into subsets which mirror the transcriptional and cytokine profile of their T cell subset counterparts. DC diversification into functional subsets provides for modules of pathogen sensing and cytokine production that direct pathogen-appropriate ILC and T cell responses. This review focuses on the recent advances in the understanding of DC development, and their function in orchestrating the innate immune modules.
- Published
- 2014
34. MONDAY PUSHLIVE TEST L-Myc expression by dendritic cells is required for optimal T-cell priming
- Author
-
Vivek Durai, Theresa L. Murphy, Marco Colonna, Emilie V. Russler-Germain, Barry P. Sleckman, Michel C. Nussenzweig, Ansuman T. Satpathy, Aaron S. Rapaport, Stephen P. Persaud, Jakob Loschko, Marina Cella, Kenneth M. Murphy, Jörn C. Albring, Nicole M. Kretzer, Paul M. Allen, Brian T. Edelson, Xiaodi Wu, Carlos G. Briseño, and Wumesh Kc
- Subjects
Multidisciplinary ,medicine.anatomical_structure ,T cell ,medicine ,Priming (immunology) ,Biology ,Cell biology - Published
- 2014
- Full Text
- View/download PDF
35. Zeb2 is required for development of plasmacytoid dendritic cells
- Author
-
Xiaodi Wu, Carlos G. Briseño, Gary E. Grajales-Reyes, KC Wumesh, Nicole M. Kretzer, Theresa L. Murphy, and Kenneth M. Murphy
- Subjects
Immunology ,Immunology and Allergy - Abstract
Recent work has shown that the transcription factor Zeb2 is regulated by and cooperates with T-bet to promote terminal differentiation of NK cells and cytotoxic T cells. In the mouse, Zeb2 is expressed throughout dendritic cell (DC) development and in mature plasmacytoid DCs (pDCs) and CD11b+ classical DCs (cDCs), but not in CD8+/CD103+ cDCs. We hypothesized that downregulation of Zeb2 expression would be essential for the development of CD8+/CD103+ cDCs. Upon conditional deletion of Zeb2, we observed a substantial decrease in pDC frequency and an increased ratio of CD8+:CD11b+ cDCs; meanwhile, Zeb2 haploinsufficiency yielded a partial defect in pDC development. Additional experiments demonstrated that this defect was cell-intrinsic and durable, and that it could not be rescued by administration of type I interferon, which increased pDC frequency in cultures of wild-type DC progenitors but directed Zeb2-deficient DC progenitors almost entirely to the CD8+/CD103+ cDC lineage. Deletion of Zeb2 induced by poly(I:C) treatment in an Mx1-Cre-driven model also resulted in perturbations in other myeloid lineages. Although poly(I:C) treatment led to an acute loss of Ly-6Chi monocytes both in Zeb2fl/fl;Mx1-Cre(tg) mice and in control mice, those monocytes were replenished to baseline frequencies in control mice but not in Zeb2-deficient mice. Similar decreases in monocyte frequency were observed in other organs, and further experiments suggested that this defect was also cell-intrinsic, gene dosage-dependent, durable, and observable in model systems that do not require use of poly(I:C). Thus, we have identified an essential regulator of development in multiple myeloid lineages.
- Published
- 2016
- Full Text
- View/download PDF
36. L-Myc expression by dendritic cells is required for optimal T-cell priming
- Author
-
Vivek Durai, Brian T. Edelson, Kenneth M. Murphy, Barry P. Sleckman, Carlos G. Briseño, Ansuman T. Satpathy, Marco Colonna, Marina Cella, Paul M. Allen, Jörn C. Albring, Wumesh Kc, Emilie V. Russler-Germain, Nicole M. Kretzer, Stephen P. Persaud, Michel C. Nussenzweig, Xiaodi Wu, Jakob Loschko, Theresa L. Murphy, and Aaron S. Rapaport
- Subjects
Male ,Transcription, Genetic ,T cell ,T-Lymphocytes ,Priming (immunology) ,Article ,Proto-Oncogene Proteins c-myc ,Mice ,Immune system ,Cross-Priming ,Antigens, CD ,medicine ,Animals ,Transcription factor ,Lung ,Regulation of gene expression ,Inflammation ,Multidisciplinary ,biology ,Cell growth ,Granulocyte-Macrophage Colony-Stimulating Factor ,Dendritic Cells ,Vesiculovirus ,biology.organism_classification ,Listeria monocytogenes ,Cell biology ,medicine.anatomical_structure ,Gene Expression Regulation ,Liver ,Vesicular stomatitis virus ,Immunology ,Interferon Regulatory Factors ,Female ,IRF8 ,Integrin alpha Chains ,Cell Division - Abstract
The transcription factors c-Myc and N-Myc--encoded by Myc and Mycn, respectively--regulate cellular growth and are required for embryonic development. A third paralogue, Mycl1, is dispensable for normal embryonic development but its biological function has remained unclear. To examine the in vivo function of Mycl1 in mice, we generated an inactivating Mycl1(gfp) allele that also reports Mycl1 expression. We find that Mycl1 is selectively expressed in dendritic cells (DCs) of the immune system and controlled by IRF8, and that during DC development, Mycl1 expression is initiated in the common DC progenitor concurrent with reduction in c-Myc expression. Mature DCs lack expression of c-Myc and N-Myc but maintain L-Myc expression even in the presence of inflammatory signals such as granulocyte-macrophage colony-stimulating factor. All DC subsets develop in Mycl1-deficient mice, but some subsets such as migratory CD103(+) conventional DCs in the lung and liver are greatly reduced at steady state. Importantly, loss of L-Myc by DCs causes a significant decrease in in vivo T-cell priming during infection by Listeria monocytogenes and vesicular stomatitis virus. The replacement of c-Myc by L-Myc in immature DCs may provide for Myc transcriptional activity in the setting of inflammation that is required for optimal T-cell priming.
- Published
- 2012
37. Heme-Mediated SPI-C Induction Promotes Monocyte Differentiation into Iron-Recycling Macrophages
- Author
-
Namakkal S. Rajasekaran, Hisashi Arase, Li Wang, Wumesh Kc, Carlos G. Briseño, Kazuhiko Igarashi, Malay Haldar, David Baltimore, Alex Yick Lun So, Nicole M. Kretzer, Ansuman T. Satpathy, Xiaodi Wu, Kenneth M. Murphy, Theresa L. Murphy, Masako Kohyama, and Takeshi Egawa
- Subjects
Male ,Mice, 129 Strain ,Cellular differentiation ,Iron ,Spleen ,Bone Marrow Cells ,Heme ,Biology ,DNA-binding protein ,General Biochemistry, Genetics and Molecular Biology ,Article ,Monocytes ,chemistry.chemical_compound ,Mice ,medicine ,Animals ,Myeloid Cells ,Transcription factor ,Derepression ,Biochemistry, Genetics and Molecular Biology(all) ,Macrophages ,Cell Differentiation ,equipment and supplies ,Molecular biology ,Spic ,DNA-Binding Proteins ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Basic-Leucine Zipper Transcription Factors ,chemistry ,Monocyte differentiation ,Female - Abstract
SummarySplenic red pulp macrophages (RPM) degrade senescent erythrocytes and recycle heme-associated iron. The transcription factor SPI-C is selectively expressed by RPM and is required for their development, but the physiologic stimulus inducing Spic is unknown. Here, we report that Spic also regulated the development of F4/80+VCAM1+ bone marrow macrophages (BMM) and that Spic expression in BMM and RPM development was induced by heme, a metabolite of erythrocyte degradation. Pathologic hemolysis induced loss of RPM and BMM due to excess heme but induced Spic in monocytes to generate new RPM and BMM. Spic expression in monocytes was constitutively inhibited by the transcriptional repressor BACH1. Heme induced proteasome-dependent BACH1 degradation and rapid Spic derepression. Furthermore, cysteine-proline dipeptide motifs in BACH1 that mediate heme-dependent degradation were necessary for Spic induction by heme. These findings are the first example of metabolite-driven differentiation of a tissue-resident macrophage subset and provide new insights into iron homeostasis.
- Full Text
- View/download PDF
38. Regulation of monocyte cell fate by blood vessels mediated by Notch signalling
- Author
-
Jaba Gamrekelashvili, Roberto Giagnorio, Jasmin Jussofie, Oliver Soehnlein, Johan Duchene, Carlos G. Briseño, Saravana K. Ramasamy, Kashyap Krishnasamy, Anne Limbourg, Christine Häger, Tamar Kapanadze, Chieko Ishifune, Rabea Hinkel, Freddy Radtke, Lothar J. Strobl, Ursula Zimber-Strobl, L. Christian Napp, Johann Bauersachs, Hermann Haller, Koji Yasutomo, Christian Kupatt, Kenneth M. Murphy, Ralf H. Adams, Christian Weber, Florian P. Limbourg, Biochemie, RS: CARIM - R3.07 - Structure-function analysis of the chemokine interactome for therapeutic targeting and imaging in atherosclerosis, RS: CARIM - R1.01 - Blood proteins & engineering, and Pathology
- Subjects
0301 basic medicine ,Male ,Myeloid ,Science ,Population ,Notch signaling pathway ,General Physics and Astronomy ,Spleen ,Bone Marrow Cells ,Cell fate determination ,Biology ,GPI-Linked Proteins ,General Biochemistry, Genetics and Molecular Biology ,Monocytes ,03 medical and health sciences ,Mice ,In vivo ,MD Multidisciplinary ,medicine ,Animals ,Antigens, Ly ,Humans ,Receptor, Notch2 ,education ,Cells, Cultured ,Mice, Knockout ,education.field_of_study ,Multidisciplinary ,Monocyte ,Calcium-Binding Proteins ,Receptors, IgG ,Endothelial Cells ,Cell Differentiation ,General Chemistry ,Adoptive Transfer ,Corrigenda ,Healthy Volunteers ,Recombinant Proteins ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Intercellular Signaling Peptides and Proteins ,Bone marrow ,Signal Transduction - Abstract
A population of monocytes, known as Ly6Clo monocytes, patrol blood vessels by crawling along the vascular endothelium. Here we show that endothelial cells control their origin through Notch signalling. Using combinations of conditional genetic deletion strategies and cell-fate tracking experiments we show that Notch2 regulates conversion of Ly6Chi monocytes into Ly6Clo monocytes in vivo and in vitro, thereby regulating monocyte cell fate under steady-state conditions. This process is controlled by Notch ligand delta-like 1 (Dll1) expressed by a population of endothelial cells that constitute distinct vascular niches in the bone marrow and spleen in vivo, while culture on recombinant DLL1 induces monocyte conversion in vitro. Thus, blood vessels regulate monocyte conversion, a form of committed myeloid cell fate regulation.
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.