26 results on '"Juliana Idoyaga"'
Search Results
2. Landscape of coordinated immune responses to H1N1 challenge in humans
- Author
-
Shannon R. Christensen, Garry P. Nolan, Nima Aghaeepour, Nilanjan Mukherjee, Sizun Jiang, Zainab Rahil, Han Chen, Jennifer Yo, Zach Bjornson-Hooper, Salil S. Bhate, Brice Gaudilliere, Matthew H. Spitzer, David R. McIlwain, Gabriela K. Fragiadakis, Pier Federico Gherardini, Rebecca Leylek, Nelson Kim, Scott E. Hensley, Kenneth Kim, Christian M. Schürch, Bonnie Bock, Juliana Idoyaga, and Melton Affrime
- Subjects
0301 basic medicine ,Male ,T-Lymphocytes ,Adaptive immunity ,Immunology ,Plasmacytoid dendritic cell ,Biology ,Lymphocyte Activation ,Virus ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Influenza A Virus, H1N1 Subtype ,Influenza, Human ,medicine ,Humans ,Viral shedding ,B cell ,Innate immunity ,Infectious disease ,Innate immune system ,Settore BIO/11 ,Monocyte ,General Medicine ,Acquired immune system ,Influenza ,Killer Cells, Natural ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Female ,Research Article - Abstract
Influenza is a significant cause of morbidity and mortality worldwide. Here we show changes in the abundance and activation states of more than 50 immune cell subsets in 35 individuals over 11 time points during human A/California/2009 (H1N1) virus challenge monitored using mass cytometry along with other clinical assessments. Peak change in monocyte, B cell, and T cell subset frequencies coincided with peak virus shedding, followed by marked activation of T and NK cells. Results led to the identification of CD38 as a critical regulator of plasmacytoid dendritic cell function in response to influenza virus. Machine learning using study-derived clinical parameters and single-cell data effectively classified and predicted susceptibility to infection. The coordinated immune cell dynamics defined in this study provide a framework for identifying novel correlates of protection in the evaluation of future influenza therapeutics.
- Published
- 2020
3. Nursing Markedly Protects Postpartum Mice From Stroke: Associated Central and Peripheral Neuroimmune Changes and a Role for Oxytocin
- Author
-
Creed M. Stary, Lijun Xu, Ludmilla A. Voloboueva, Marcela Alcántara-Hernández, Oiva J. Arvola, Juliana Idoyaga, and Rona G. Giffard
- Subjects
0301 basic medicine ,FACS ,Ischemia ,Spleen ,Inflammation ,ischemia ,lcsh:RC321-571 ,03 medical and health sciences ,0302 clinical medicine ,Nursing ,medicine ,innate ,cytokine ,Stroke ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,Neuroinflammation ,Original Research ,business.industry ,General Neuroscience ,adaptive ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,Oxytocin ,inflammation ,focal ischemia ,medicine.symptom ,MCAO ,business ,030217 neurology & neurosurgery ,Postpartum period ,medicine.drug ,Hormone ,Neuroscience - Abstract
Recent studies demonstrate significant neuroimmune changes in postpartum females, a period that also carries an increased risk of stroke. Oxytocin, a major hormone upregulated in the brains of nursing mothers, has been shown to both modulate neuroinflammation and protect against stroke. In the present study we assessed whether and how nursing modulates the neuroimmune response and injury after stroke. We observed that postpartum nursing mice were markedly protected from 1 h of transient middle cerebral artery occlusion (MCAO) relative to either non-pregnant/non-postpartum or non-nursing (pups removed) postpartum females. Nursing mice also expressed reduced levels of pro-inflammatory cytokines, had decreased migration of blood leukocytes into the brain following MCAO, and displayed peripheral neuroimmune changes characterized by increased spleen weight and increased fraction of spleen monocytes. Intranasal oxytocin treatment in non-pregnant females in part recapitulated the protective and anti-inflammatory effects associated with nursing. In summary, the results of the present study demonstrate that nursing in the postpartum period provides relative protection against transient ischemic stroke associated with decreased brain leukocytes and increased splenic monocytes. These effects appear to be regulated, at least in part, by oxytocin.
- Published
- 2019
4. The versatile plasmacytoid dendritic cell: Function, heterogeneity, and plasticity
- Author
-
Juliana Idoyaga and Rebecca Leylek
- Subjects
0303 health sciences ,030302 biochemistry & molecular biology ,Cell ,Antigen presentation ,hemic and immune systems ,Plasmacytoid dendritic cell ,Biology ,medicine.disease_cause ,Autoimmunity ,03 medical and health sciences ,medicine.anatomical_structure ,Immune system ,Interferon ,medicine ,Compartment (development) ,Neuroscience ,Function (biology) ,medicine.drug - Abstract
Since their identification as the natural interferon-producing cell two decades ago, plasmacytoid dendritic cells (pDCs) have been attributed diverse functions in the immune response. Their most well characterized function is innate, i.e., their rapid and robust production of type-I interferon (IFN-I) in response to viruses. However, pDCs have also been implicated in antigen presentation, activation of adaptive immune responses and immunoregulation. The mechanisms by which pDCs enact these diverse functions are poorly understood. One central debate is whether these functions are carried out by different pDC subpopulations or by plasticity in the pDC compartment. This chapter summarizes the latest reports regarding pDC function, heterogeneity, cell conversion and environmentally influenced plasticity, as well as the role of pDCs in infection, autoimmunity and cancer.
- Published
- 2019
- Full Text
- View/download PDF
5. Monocyte-derived dendritic cells enhance protection against secondary influenza challenge by controlling the switch in CD8+ T-cell immunodominance
- Author
-
Paula Ruibal, Juliana Idoyaga, César Muñoz-Fontela, Anja Lüdtke, Sergio Gómez-Medina, Jazmina L. G. Cruz, and José Vicente Pérez-Girón
- Subjects
0301 basic medicine ,Monocyte ,Immunology ,Dendritic cell ,Immunodominance ,Biology ,Virology ,Virus ,Nucleoprotein ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,medicine ,biology.protein ,Immunology and Allergy ,Cytotoxic T cell ,CD8 ,Polymerase - Abstract
Influenza virus infection triggers an increase in the number of monocyte-derived dendritic cells (moDCs) in the respiratory tract, but the role of these cells during antiviral immunity is still unclear. Here we show that during influenza infection, moDCs dominate the late activation of CD8+ T cells and trigger the switch in immunodominance of the CD8+ T-cell response from acidic polymerase specificity to nucleoprotein specificity. Abrogation of monocyte recruitment or depletion of moDCs strongly compromised host resistance to secondary influenza challenge. These findings underscore a novel function of moDCs in the antiviral response to influenza virus, and have important implications for vaccine design.
- Published
- 2016
- Full Text
- View/download PDF
6. Expansion and Activation of CD103+ Dendritic Cell Progenitors at the Tumor Site Enhances Tumor Responses to Therapeutic PD-L1 and BRAF Inhibition
- Author
-
Brian D. Brown, Romain Remark, Florent Ginhoux, Sacha Gnjatic, Judith Agudo, Marcus Bosenberg, Juliana Idoyaga, Anna Karolina Palucka, Daigo Hashimoto, Stefan Jordan, Maria Casanova-Acebes, Nina Bhardwaj, Marylene Leboeuf, Christina Rivera, Adeeb Rahman, Svetoslav Chakarov, Joshua Brody, Makhzuna Khudoynazarova, Miriam Merad, Navpreet Tung, Hélène Salmon, and Brandon Hogstad
- Subjects
Proto-Oncogene Proteins B-raf ,0301 basic medicine ,Immunology ,Melanoma, Experimental ,Priming (immunology) ,chemical and pharmacologic phenomena ,CD8-Positive T-Lymphocytes ,B7-H1 Antigen ,03 medical and health sciences ,0302 clinical medicine ,Antigens, CD ,Cell Line, Tumor ,PD-L1 ,medicine ,Animals ,Immunology and Allergy ,Progenitor cell ,Mice, Knockout ,Antigen Presentation ,biology ,Melanoma ,hemic and immune systems ,Dendritic Cells ,Dendritic cell ,medicine.disease ,3. Good health ,Blockade ,Mice, Inbred C57BL ,Poly I-C ,030104 developmental biology ,Infectious Diseases ,fms-Like Tyrosine Kinase 3 ,030220 oncology & carcinogenesis ,biology.protein ,Systemic administration ,Cancer research ,Integrin alpha Chains ,CD8 - Abstract
Large numbers of melanoma lesions develop resistance to targeted inhibition of mutant BRAF or fail to respond to checkpoint blockade. We explored whether modulation of intratumoral antigen-presenting cells (APCs) could increase responses to these therapies. Using mouse melanoma models, we found that CD103(+) dendritic cells (DCs) were the only APCs transporting intact antigens to the lymph nodes and priming tumor-specific CD8(+) T cells. CD103(+) DCs were required to promote anti-tumoral effects upon blockade of the checkpoint ligand PD-L1; however, PD-L1 inhibition only led to partial responses. Systemic administration of the growth factor FLT3L followed by intratumoral poly I:C injections expanded and activated CD103(+) DC progenitors in the tumor, enhancing responses to BRAF and PD-L1 blockade and protecting mice from tumor rechallenge. Thus, the paucity of activated CD103(+) DCs in tumors limits checkpoint-blockade efficacy and combined FLT3L and poly I:C therapy can enhance tumor responses to checkpoint and BRAF blockade.
- Published
- 2016
- Full Text
- View/download PDF
7. The Nontoxic Cholera B Subunit Is a Potent Adjuvant for Intradermal DC-Targeted Vaccination
- Author
-
Laura Antonio-Herrera, Oscar Badillo-Godinez, Oscar Medina-Contreras, Araceli Tepale-Segura, Alberto García-Lozano, Lourdes Gutierrez-Xicotencatl, Gloria Soldevila, Fernando R. Esquivel-Guadarrama, Juliana Idoyaga, and Laura C. Bonifaz
- Subjects
Male ,Rotavirus ,0301 basic medicine ,anti-DEC205 ,Cellular differentiation ,medicine.medical_treatment ,Priming (immunology) ,Lymphocyte Activation ,memory ,Mice ,Immunology and Allergy ,Receptor ,Melanoma ,Original Research ,Mice, Inbred BALB C ,biology ,Vaccination ,3. Good health ,Treatment Outcome ,medicine.anatomical_structure ,Female ,Antibody ,Adjuvant ,lcsh:Immunologic diseases. Allergy ,Cholera Toxin ,skin ,Injections, Intradermal ,T cell ,Immunology ,T cells ,Mice, Transgenic ,Receptors, Cell Surface ,complex mixtures ,Rotavirus Infections ,Minor Histocompatibility Antigens ,03 medical and health sciences ,Immune system ,Adjuvants, Immunologic ,Antigen ,adjuvant ,Antigens, CD ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Lectins, C-Type ,dendritic cells ,business.industry ,Th1 Cells ,CTB ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,biology.protein ,Th17 Cells ,business ,lcsh:RC581-607 - Abstract
CD4+ T cells are major players in the immune response against several diseases; including AIDS, leishmaniasis, tuberculosis, influenza and cancer. Their activation has been successfully achieved by administering antigen coupled with antibodies, against DC-specific receptors in combination with adjuvants. Unfortunately, most of the adjuvants used so far in experimental models are unsuitable for human use. Therefore, human DC-targeted vaccination awaits the description of potent, yet nontoxic adjuvants. The nontoxic cholera B subunit (CTB) can be safely used in humans and it has the potential to activate CD4+ T cell responses. However, it remains unclear whether CTB can promote DC activation and can act as an adjuvant for DC-targeted antigens. Here, we evaluated the CTB's capacity to activate DCs and CD4+ T cell responses, and to generate long-lasting protective immunity. Intradermal (i.d.) administration of CTB promoted late and prolonged activation and accumulation of skin and lymphoid-resident DCs. When CTB was co-administered with anti-DEC205-OVA, it promoted CD4+ T cell expansion, differentiation, and infiltration to peripheral nonlymphoid tissues, i.e., the skin, lungs and intestine. Indeed, CTB promoted a polyfunctional CD4+ T cell response, including the priming of Th1 and Th17 cells, as well as resident memory T (RM) cell differentiation in peripheral nonlymphoid tissues. It is worth noting that CTB together with a DC-targeted antigen promoted local and systemic protection against experimental melanoma and murine rotavirus. We conclude that CTB administered i.d. can be used as an adjuvant to DC-targeted antigens for the induction of broad CD4+ T cell responses as well as for promoting long-lasting protective immunity.
- Published
- 2018
- Full Text
- View/download PDF
8. Vaccination-induced skin-resident memory CD8+ T cells mediate strong protection against cutaneous melanoma
- Author
-
Felipe Gálvez-Cancino, Juliana Idoyaga, Flavio Salazar-Onfray, Ornella Chovar, Pablo Cáceres, Ernesto Lopez, Evelyn Menares, Ximena Díaz, Manuel Varas-Godoy, Marcela Alcántara-Hernández, Vincenzo Borgna, Sofia Hidalgo, Alvaro Lladser, and Camila Flores
- Subjects
lcsh:Immunologic diseases. Allergy ,0301 basic medicine ,T cell ,intradermal vaccination ,Immunology ,Cell ,Biology ,Cancer vaccines ,lcsh:RC254-282 ,DNA vaccination ,DNA vaccines ,03 medical and health sciences ,0302 clinical medicine ,Immunity ,medicine ,melanoma ,tissue resident memory CD8+ T cells ,Immunology and Allergy ,Cytotoxic T cell ,Original Research ,Melanoma ,protein vaccines ,models of anticancer vaccination ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,Acquired immune system ,3. Good health ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,lcsh:RC581-607 ,CD8 - Abstract
Memory CD8+ T cell responses have the potential to mediate long-lasting protection against cancers. Resident memory CD8+ T (Trm) cells stably reside in non-lymphoid tissues and mediate superior innate and adaptive immunity against pathogens. Emerging evidence indicates that Trm cells develop in human solid cancers and play a key role in controlling tumor growth. However, the specific contribution of Trm cells to anti-tumor immunity is incompletely understood. Moreover, clinically applicable vaccination strategies that efficiently establish Trm cell responses remain largely unexplored and are expected to strongly protect against tumors. Here we demonstrated that a single intradermal administration of gene- or protein-based vaccines efficiently induces specific Trm cell responses against models of tumor-specific and self-antigens, which accumulated in vaccinated and distant non-vaccinated skin. Vaccination-induced Trm cells were largely resistant to in vivo intravascular staining and antibody-dependent depletion. Intradermal, but not intraperitoneal vaccination, generated memory precursors expressing skin-homing molecules in circulation and Trm cells in skin. Interestingly, vaccination-induced Trm cell responses strongly suppressed the growth of B16F10 melanoma, independently of circulating memory CD8+ T cells, and were able to infiltrate tumors. This work highlights the therapeutic potential of vaccination-induced Trm cell responses to achieve potent protection against skin malignancies.
- Published
- 2018
9. CDKN1A regulates Langerhans cell survival and promotes Treg cell generation upon exposure to ionizing irradiation
- Author
-
Carolina L. Bigarella, Hélène Salmon, Brandon Hogstad, Saghi Ghaffari, Jeremy Price, Juliana Idoyaga, Miriam Merad, and Marylene Leboeuf
- Subjects
0303 health sciences ,Langerhans cell ,integumentary system ,medicine.diagnostic_test ,Microarray analysis techniques ,Kinase ,DNA damage ,Immunology ,hemic and immune systems ,chemical and pharmacologic phenomena ,Biology ,3. Good health ,Flow cytometry ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Downregulation and upregulation ,Apoptosis ,medicine ,Cancer research ,Immunology and Allergy ,Lymph ,030304 developmental biology ,030215 immunology - Abstract
Treatment with ionizing radiation (IR) can lead to the accumulation of tumor-infiltrating regulatory T cells (Treg cells) and subsequent resistance of tumors to radiotherapy. Here we focused on the contribution of the epidermal mononuclear phagocytes Langerhans cells (LCs) to this phenomenon because of their ability to resist depletion by high-dose IR. We found that LCs resisted apoptosis and rapidly repaired DNA damage after exposure to IR. In particular, we found that the cyclin-dependent kinase inhibitor CDKN1A (p21) was overexpressed in LCs and that Cdkn1a(-/-) LCs underwent apoptosis and accumulated DNA damage following IR treatment. Wild-type LCs upregulated major histocompatibility complex class II molecules, migrated to the draining lymph nodes and induced an increase in Treg cell numbers upon exposure to IR, but Cdkn1a(-/-) LCs did not. Our findings suggest a means for manipulating the resistance of LCs to IR to enhance the response of cutaneous tumors to radiotherapy.
- Published
- 2015
- Full Text
- View/download PDF
10. High-Dimensional Phenotypic Mapping of Human Dendritic Cells Reveals Interindividual Variation and Tissue Specialization
- Author
-
Garry P. Nolan, Edgar G. Engleman, M. Peter Marinkovich, Mark M. Davis, Tibor Keler, Lisa E. Wagar, Juliana Idoyaga, Marcela Alcántara-Hernández, and Rebecca Leylek
- Subjects
0301 basic medicine ,Palatine Tonsil ,Gene Expression ,Transcriptome ,Variable Expression ,Mice ,0302 clinical medicine ,Neoplasms ,Immunology and Allergy ,Molecular Targeted Therapy ,Receptors, Immunologic ,Receptor ,Skin ,Biological Variation, Individual ,Antibodies, Monoclonal ,Phenotype ,Cell biology ,Infectious Diseases ,medicine.anatomical_structure ,Organ Specificity ,030220 oncology & carcinogenesis ,Female ,Cytophotometry ,Immunotherapy ,medicine.drug_class ,Immunology ,Spleen ,Biology ,Monoclonal antibody ,Cancer Vaccines ,Article ,Immunophenotyping ,03 medical and health sciences ,Antigens, CD ,Proto-Oncogene Proteins ,medicine ,Animals ,Humans ,Genetic heterogeneity ,Receptor Protein-Tyrosine Kinases ,Dendritic Cells ,Antigens, Differentiation ,Axl Receptor Tyrosine Kinase ,Mice, Inbred C57BL ,030104 developmental biology ,Tonsil ,Lymph Nodes ,Biomarkers - Abstract
Given the limited efficacy of clinical approaches that rely on ex vivo generated dendritic cells (DCs), it is imperative to design strategies that harness specialized DC subsets in situ. This requires delineating the expression of surface markers by DC subsets among individuals and tissues. Here, we performed a multiparametric phenotypic characterization and unbiased analysis of human DC subsets in blood, tonsil, spleen, and skin. We uncovered previously unreported phenotypic heterogeneity of human cDC2s among individuals, including variable expression of functional receptors such as CD172a. We found marked differences in DC subsets localized in blood and lymphoid tissues versus skin, and a striking absence of the newly discovered Axl+ DCs in the skin. Finally, we evaluated the capacity of anti-receptor monoclonal antibodies to deliver vaccine components to skin DC subsets. These results offer a promising path for developing DC subset-specific immunotherapies that cannot be provided by transcriptomic analysis alone.
- Published
- 2017
11. Ebola virus infection kinetics in chimeric mice reveal a key role of T cells as barriers for virus dissemination
- Author
-
César Muñoz-Fontela, Estefanía Rodríguez, Stephanie Wurr, David M. Wozniak, Stephan Günther, Susanne Krasemann, Lisa Oestereich, Paula Ruibal, Sabrina Bockholt, Juliana Idoyaga, Anja Lüdtke, Xiangguo Qiu, Elisa Pallasch, Sergio Gómez-Medina, and Gary P. Kobinger
- Subjects
0301 basic medicine ,Lymphoid Tissue ,T-Lymphocytes ,viruses ,Viremia ,Biology ,Virus Replication ,medicine.disease_cause ,Article ,Virus ,03 medical and health sciences ,Cross-Priming ,Antigen ,Antigens, CD ,Immunity ,Viral entry ,medicine ,Animals ,Mice, Knockout ,CD11b Antigen ,Multidisciplinary ,Ebola virus ,Dendritic Cells ,Hemorrhagic Fever, Ebola ,Ebolavirus ,medicine.disease ,Virology ,Mice, Inbred C57BL ,Kinetics ,030104 developmental biology ,Viral replication ,Host-Pathogen Interactions ,Immunology ,Integrin alpha Chains ,CD8 - Abstract
Ebola virus (EBOV) causes severe systemic disease in humans and non-human primates characterized by high levels of viremia and virus titers in peripheral organs. The natural portals of virus entry are the mucosal surfaces and the skin where macrophages and dendritic cells (DCs) are primary EBOV targets. Due to the migratory properties of DCs, EBOV infection of these cells has been proposed as a necessary step for virus dissemination via draining lymph nodes and blood. Here we utilize chimeric mice with competent hematopoietic-driven immunity, to show that EBOV primarily infects CD11b+ DCs in non-lymphoid and lymphoid tissues, but spares the main cross-presenting CD103+ DC subset. Furthermore, depletion of CD8 and CD4 T cells resulted in loss of early control of virus replication, viremia and fatal Ebola virus disease (EVD). Thus, our findings point out at T cell function as a key determinant of EVD progress and outcome.
- Published
- 2017
- Full Text
- View/download PDF
12. Integrated Cross-Species Analysis Identifies a Conserved Transitional Dendritic Cell Population
- Author
-
Zachary Lanzar, Juliana Idoyaga, Anja Lüdtke, Marcela Alcántara-Hernández, Rebecca Leylek, Oriana A. Perez, and Boris Reizis
- Subjects
0301 basic medicine ,Adult ,Male ,Population ,Antigen presentation ,Biology ,Viral infection ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Interferon-gamma ,Mice ,0302 clinical medicine ,Immune system ,Species Specificity ,Interferon ,medicine ,Animals ,Humans ,Mass cytometry ,education ,lcsh:QH301-705.5 ,Cells, Cultured ,Cyclin-dependent kinase 1 ,education.field_of_study ,Antigen Presentation ,Mice, Inbred BALB C ,hemic and immune systems ,Dendritic cell ,Dendritic Cells ,Flow Cytometry ,Orthomyxoviridae ,Mice, Inbred C57BL ,030104 developmental biology ,lcsh:Biology (General) ,Female ,Single-Cell Analysis ,Transcriptome ,Neuroscience ,030217 neurology & neurosurgery ,medicine.drug - Abstract
SUMMARY Plasmacytoid dendritic cells (pDCs) are sensor cells with diverse immune functions, from type I interferon (IFN-I) production to antigen presentation, T cell activation, and tolerance. Regulation of these functions remains poorly understood but could be mediated by functionally specialized pDC subpopulations. We address pDC diversity using a high-dimensional single-cell approach: mass cytometry (CyTOF). Our analysis uncovers a murine pDC-like population that specializes in antigen presentation with limited capacity for IFN-I production. Using a multifaceted cross-species comparison, we show that this pDC-like population is the definitive murine equivalent of the recently described human AXL+ DCs, which we unify under the name transitional DCs (tDCs) given their continuum of pDC and cDC2 characteristics. tDCs share developmental traits with pDCs, as well as recruitment dynamics during viral infection. Altogether, we provide a framework for deciphering the function of pDCs and tDCs during diseases, which has the potential to open new avenues for therapeutic design., Graphical Abstract, In Brief Dendritic cells (DCs) are unique therapeutic targets given their capacity to modulate immune responses. Yet complete alignment of the DC network between species is lacking. Using a multidimensional approach, Leylek et al. identify the mouse homolog of human AXL+ DCs, named transitional DCs (tDCs), and reveal their similarities with pDCs.
- Published
- 2019
13. Pseudogenization of the Secreted Effector Gene sseI Confers Rapid Systemic Dissemination of S. Typhimurium ST313 within Migratory Dendritic Cells
- Author
-
Trung H.M. Pham, Kyler A. Lugo, Donna M. Bouley, Renée M. Tsolis, Jared Honeycutt, Denise M. Monack, Gregory T. Walker, Amanda Jacobson, Sarah Carden, and Juliana Idoyaga
- Subjects
0301 basic medicine ,Male ,Salmonella typhimurium ,pseudogene ,Bacteremia ,Inbred C57BL ,Genome ,iNTS ,Mice ,Cell Movement ,Receptors ,Mesenteric lymph nodes ,2.1 Biological and endogenous factors ,2.2 Factors relating to the physical environment ,Aetiology ,Genetics ,Mice, Knockout ,Effector ,pathogenesis ,Bacterial ,Hematology ,Foodborne Illness ,Phenotype ,Gastroenteritis ,medicine.anatomical_structure ,T3SS effectors ,Infectious Diseases ,Medical Microbiology ,Host-Pathogen Interactions ,Salmonella Infections ,Female ,Host adaptation ,Infection ,NTS ,nontyphoidal Salmonella ,Receptors, CCR7 ,Lineage (genetic) ,Knockout ,genome degradation ,030106 microbiology ,Immunology ,Biology ,Communicable Diseases ,Microbiology ,Article ,Vaccine Related ,03 medical and health sciences ,SrfH ,Virology ,Biodefense ,evolution ,medicine ,Humans ,Animals ,Gene ,niche adaptation ,Animal ,Prevention ,Biological Transport ,Dendritic Cells ,Gene Expression Regulation, Bacterial ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Emerging Infectious Diseases ,Gene Expression Regulation ,Genes ,Genes, Bacterial ,Disease Models ,Parasitology ,Lymph Nodes ,Niche adaptation ,Digestive Diseases ,CCR7 - Abstract
© 2017 Elsevier Inc. Genome degradation correlates with host adaptation and systemic disease in Salmonella. Most lineages of the S. enterica subspecies Typhimurium cause gastroenteritis in humans; however, the recently emerged ST313 lineage II pathovar commonly causes systemic bacteremia in sub-Saharan Africa. ST313 lineage II displays genome degradation compared to gastroenteritis-associated lineages; yet, the mechanisms and causal genetic differences mediating these infection phenotypes are largely unknown. We find that the ST313 isolate D23580 hyperdisseminates from the gut to systemic sites, such as the mesenteric lymph nodes (MLNs), via CD11b+ migratory dendritic cells (DCs). This hyperdissemination was facilitated by the loss of sseI, which encodes an effector that inhibits DC migration in gastroenteritis-associated isolates. Expressing functional SseI in D23580 reduced the number of infected migratory DCs and bacteria in the MLN. Our study reveals a mechanism linking pseudogenization of effectors with the evolution of niche adaptation in a bacterial pathogen.
- Published
- 2016
- Full Text
- View/download PDF
14. Ebola Virus Disease Is Characterized by Poor Activation and Reduced Levels of Circulating CD16 + Monocytes
- Author
-
Romy Kerber, Anja Lüdtke, César Muñoz-Fontela, Beate Becker-Ziaja, Mar Cabeza-Cabrerizo, N’Faly Magassouba, Monika Rottstegge, Paula Ruibal, David M. Wozniak, Martin Gabriel, Anja Thorenz, Lisa Oestereich, Stephan Günther, Romy Weller, Juliana Idoyaga, and TwinCore, Centre for experimental and clinical infection research GmbH, Feodor-Lynen-Str. 7, 30625 Hannover, Germany.
- Subjects
0301 basic medicine ,Neutrophils ,viruses ,CD14 ,Disease ,CD16 ,Biology ,medicine.disease_cause ,Monocytes ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,Immunology and Allergy ,Antigen-presenting cell ,Ebola virus ,Receptors, IgG ,virus diseases ,Dendritic Cells ,Hemorrhagic Fever, Ebola ,Ebolavirus ,Phenotype ,Virology ,Pathophysiology ,3. Good health ,Kinetics ,030104 developmental biology ,Infectious Diseases ,Immunology ,T cell subset ,Female ,Mobile Health Units ,030215 immunology - Abstract
A number of previous studies have identified antigen-presenting cells (APCs) as key targets of Ebola virus (EBOV), but the role of APCs in human Ebola virus disease (EVD) is not known. We have evaluated the phenotype and kinetics of monocytes, neutrophils, and dendritic cells (DCs) in peripheral blood of patients for whom EVD was diagnosed by the European Mobile Laboratory in Guinea. Acute EVD was characterized by reduced levels of circulating nonclassical CD16(+) monocytes with a poor activation profile. In survivors, CD16(+) monocytes were activated during recovery, coincident with viral clearance, suggesting an important role of this cell subset in EVD pathophysiology.
- Published
- 2016
- Full Text
- View/download PDF
15. Streamlined Expressed Protein Ligation Using Split Inteins
- Author
-
Neel H. Shah, Zhihua Liu, Juliana Idoyaga, Tom W. Muir, John A. Willis, and Miquel Vila-Perelló
- Subjects
dnaE ,Peptide ,Protein Engineering ,010402 general chemistry ,01 natural sciences ,Biochemistry ,Article ,Catalysis ,Inteins ,law.invention ,03 medical and health sciences ,Colloid and Surface Chemistry ,law ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,Chemistry ,Extramural ,Antibodies, Monoclonal ,Esters ,General Chemistry ,Protein engineering ,Recombinant Proteins ,0104 chemical sciences ,Enzyme ,Recombinant DNA ,Intein ,Ligation - Abstract
Chemically modified proteins are invaluable tools for studying the molecular details of biological processes, and they also hold great potential as new therapeutic agents. Several methods have been developed for the site-specific modification of proteins, one of the most widely used being expressed protein ligation (EPL) in which a recombinant α-thioester is ligated to an N-terminal Cys-containing peptide. Despite the widespread use of EPL, the generation and isolation of the required recombinant protein α-thioesters remain challenging. We describe here a new method for the preparation and purification of recombinant protein α-thioesters using engineered versions of naturally split DnaE inteins. This family of autoprocessing enzymes is closely related to the inteins currently used for protein α-thioester generation, but they feature faster kinetics and are split into two inactive polypeptides that need to associate to become active. Taking advantage of the strong affinity between the two split intein fragments, we devised a streamlined procedure for the purification and generation of protein α-thioesters from cell lysates and applied this strategy for the semisynthesis of a variety of proteins including an acetylated histone and a site-specifically modified monoclonal antibody.
- Published
- 2012
- Full Text
- View/download PDF
16. Expression of the zinc finger transcription factor zDC (Zbtb46, Btbd4) defines the classical dendritic cell lineage
- Author
-
Alice O. Kamphorst, Matthew M. Meredith, Guillaume Darrasse-Jèze, Pierre Guermonprez, Cheolho Cheong, Kang Liu, Kai Hui Yao, Rachel E. Niec, Michel C. Nussenzweig, Juliana Idoyaga, and Heidi A. Schreiber
- Subjects
Immunology ,CD11c ,Biology ,Article ,Monocytes ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,Immunity ,medicine ,Immunology and Allergy ,Animals ,Cell Lineage ,Diphtheria Toxin ,Progenitor cell ,Receptor ,3' Untranslated Regions ,Melanoma ,health care economics and organizations ,030304 developmental biology ,Diphtheria toxin ,Zinc finger transcription factor ,Mice, Knockout ,0303 health sciences ,Macrophages ,hemic and immune systems ,Zinc Fingers ,Dendritic Cells ,Molecular biology ,3. Good health ,CD11c Antigen ,Killer Cells, Natural ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Gene Expression Regulation ,Intercellular Signaling Peptides and Proteins ,Bone marrow ,Toxoplasma ,Toxoplasmosis ,030215 immunology ,Heparin-binding EGF-like Growth Factor ,Transcription Factors - Abstract
The zinc finger transcription factor zDC is uniquely expressed by the cDC lineage among immune cells, and the insertion of diphtheria toxin receptor cDNA into the zDC locus allows specific ablation of the cDC lineage in mice., Classical dendritic cells (cDCs), monocytes, and plasmacytoid DCs (pDCs) arise from a common bone marrow precursor (macrophage and DC progenitors [MDPs]) and express many of the same surface markers, including CD11c. We describe a previously uncharacterized zinc finger transcription factor, zDC (Zbtb46, Btbd4), which is specifically expressed by cDCs and committed cDC precursors but not by monocytes, pDCs, or other immune cell populations. We inserted diphtheria toxin (DT) receptor (DTR) cDNA into the 3′ UTR of the zDC locus to serve as an indicator of zDC expression and as a means to specifically deplete cDCs. Mice bearing this knockin express DTR in cDCs but not other immune cell populations, and DT injection into zDC-DTR bone marrow chimeras results in cDC depletion. In contrast to previously characterized CD11c-DTR mice, non-cDCs, including pDCs, monocytes, macrophages, and NK cells, were spared after DT injection in zDC-DTR mice. We compared immune responses to Toxoplasma gondii and MO4 melanoma in DT-treated zDC- and CD11c-DTR mice and found that immunity was only partially impaired in zDC-DTR mice. Our results indicate that CD11c-expressing non-cDCs make significant contributions to initiating immunity to parasites and tumors.
- Published
- 2012
17. Targeting of antigens to skin dendritic cells: possibilities to enhance vaccine efficacy
- Author
-
Juliana Idoyaga, Nikolaus Romani, Martin Thurnher, Ralph M. Steinman, Vincent Flacher, and Innsbruck Medical University [Austria] (IMU)
- Subjects
Antigen Targeting ,Langerin ,medicine.medical_treatment ,CD14 ,Immunology ,Human skin ,Article ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,Antigens, CD ,medicine ,Animals ,Humans ,Immunology and Allergy ,Cytotoxic T cell ,Lectins, C-Type ,ComputingMilieux_MISCELLANEOUS ,030304 developmental biology ,Vaccines ,0303 health sciences ,integumentary system ,biology ,Cell Biology ,Immunotherapy ,3. Good health ,Mannose-Binding Lectins ,Langerhans Cells ,biology.protein ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,Antibody ,030215 immunology - Abstract
Vaccinations in medicine are commonly administered through the skin. Therefore, the vaccine is immunologically processed by antigen-presenting cells of the skin. There is recent evidence that the clinically less often used intradermal route is effective; in cases even superior to the conventional subcutaneous or intramuscular route. Professional antigen-presenting cells of the skin comprise epidermal Langerhans cells (CD207/langerin(+)), dermal langerin(-) and dermal langerin(+) dendritic cells (DCs). In human skin, langerin(-) dermal DCs can be further subdivided on the basis of their reciprocal CD1a and CD14 expression. The relative contributions of these subsets to the generation of immunity or tolerance are still unclear. Langerhans cells in human skin seem to be specialized for induction of cytotoxic T lymphocytes. Likewise, mouse Langerhans cells are capable of cross-presentation and of protecting against experimental tumours. It is desirable to harness these properties for immunotherapy. A promising strategy to dramatically improve the outcome of vaccinations is 'antigen targeting'. Thereby, the vaccine is delivered directly and selectively to defined types of skin DCs. Targeting is achieved by means of coupling antigen to antibodies that recognize cell surface receptors on DCs. This approach is being widely explored. Little is known, however, about the events that take place in the skin and the DCs subsets involved therein. This topic will be discussed in this article.
- Published
- 2010
- Full Text
- View/download PDF
18. Innate NKT lymphocytes confer superior adaptive immunity via tumor-capturing dendritic cells
- Author
-
Juliana Idoyaga, Anthony J. Bonito, Ralph M. Steinman, Shin-ichiro Fujii, Yang Liu, Xue-Feng Bai, Rosa Wainstok, José Mordoh, Kang Liu, and Anna Charalambous
- Subjects
CD4-Positive T-Lymphocytes ,Lymphoma ,major histocompatibility antigen class 1 ,cell maturation ,animal cell ,CD8-Positive T-Lymphocytes ,Lymphocyte Activation ,Mice ,0302 clinical medicine ,antibody ,drug uptake ,purl.org/becyt/ford/3.2 [https] ,T lymphocyte ,drug delivery system ,Immunology and Allergy ,Antigen Presentation ,Mice, Inbred BALB C ,0303 health sciences ,irradiation ,article ,Cell Differentiation ,CD8 antigen ,lymph node ,Natural killer T cell ,Acquired immune system ,major histocompatibility complex ,alpha CD40 antibody ,polyinosinic polycytidylic acid ,unclassified drug ,3. Good health ,alpha galactosylceramide ,lymphoid organ ,Killer Cells, Natural ,female ,medicine.anatomical_structure ,priority journal ,Female ,purl.org/becyt/ford/3 [https] ,CD4 antigen ,dendritic cell ,animal experiment ,Immunology ,Antigen presentation ,Biology ,Cancer Vaccines ,Article ,Natural killer cell ,03 medical and health sciences ,Immune system ,adjuvant ,plasmacytoma ,Adjuvants, Immunologic ,Antigen ,Antigens, Neoplasm ,medicine ,Animals ,controlled study ,mouse ,Tumors ,030304 developmental biology ,nonhuman ,animal model ,Immunity ,Immunotherapy, Active ,Neoplasms, Experimental ,Dendritic Cells ,Dendritic cell ,natural killer cell ,vaccination ,tumor resistance ,tumor immunity ,Gamma Rays ,spleen ,Lymph Nodes ,glycolipid ,Spleen ,tumor antigen ,Neoplasm Transplantation ,CD8 ,Plasmacytoma ,spleen cell ,030215 immunology - Abstract
If irradiated tumor cells could be rendered immunogenic, they would provide a safe, broad, and patient-specific array of antigens for immunotherapies. Prior approaches have emphasized genetic transduction of live tumor cells to express cytokines, costimulators, and surrogate foreign antigens. We asked if immunity could be achieved by delivering irradiated, major histocompatibility complex-negative plasmacytoma cells to maturing mouse dendritic cells (DCs) within lymphoid organs. Tumor cells injected intravenously (i.v.) were captured by splenic DCs, whereas subcutaneous (s.c.) injection led only to weak uptake in lymph node or spleen. The natural killer T (NKT) cells mobilizing glycolipid α-galactosyl ceramide, used to mature splenic DCs, served as an effective adjuvant to induce protective immunity. This adjuvant function was mimicked by a combination of poly IC and agonistic αCD40 antibody. The adjuvant glycolipid had to be coadministered with tumor cells i.v. rather than s.c. Specific resistance was generated both to a plasmacytoma and lymphoma. The resistance afforded by a single vaccination lasted >2 mo and required both CD4+ and CD8+ T cells. Mature tumor capturing DCs stimulated the differentiation of P1A tumor antigen-specific, CD8+ T cells and uniquely transferred tumor resistance to naive mice. Therefore, the access of dying tumor cells to DCs that are maturing to activated NKT cells efficiently induces long-lived adaptive resistance. Fil: Liu, Kang. The Rockefeller University; Estados Unidos Fil: Idoyaga, Juliana. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Parque Centenario. Instituto de Investigaciones Bioquímicas de Buenos Aires. Fundación Instituto Leloir. Instituto de Investigaciones Bioquímicas de Buenos Aires; Argentina Fil: Charalambous, Anna. The Rockefeller University; Estados Unidos Fil: Fujii, Shin Ichiro. The Rockefeller University; Estados Unidos Fil: Bonito, Anthony. The Rockefeller University; Estados Unidos Fil: Mordoh, Jose. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Parque Centenario. Instituto de Investigaciones Bioquímicas de Buenos Aires. Fundación Instituto Leloir. Instituto de Investigaciones Bioquímicas de Buenos Aires; Argentina Fil: Wainstok, Rosa. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Parque Centenario. Instituto de Investigaciones Bioquímicas de Buenos Aires. Fundación Instituto Leloir. Instituto de Investigaciones Bioquímicas de Buenos Aires; Argentina. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Ciudad Universitaria. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales. Universidad de Buenos Aires. Facultad de Ciencias Exactas y Naturales. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales; Argentina Fil: Bai, Xue Feng. Ohio State University; Estados Unidos Fil: Liu, Yang. Ohio State University; Estados Unidos Fil: Steinman, Ralph M.. The Rockefeller University; Estados Unidos
- Published
- 2005
- Full Text
- View/download PDF
19. Reply to: 'Subverting misconceptions about radiation therapy'
- Author
-
Miriam Merad, Jeremy Price, and Juliana Idoyaga
- Subjects
Cyclin-Dependent Kinase Inhibitor p21 ,0301 basic medicine ,medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Immunology ,T-Lymphocytes, Regulatory ,Radiation therapy ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Langerhans Cells ,Radiation, Ionizing ,030220 oncology & carcinogenesis ,Animals ,Immunology and Allergy ,Medicine ,Medical physics ,business - Published
- 2016
- Full Text
- View/download PDF
20. BRAF-V600E expression in precursor versus differentiated dendritic cells defines clinically distinct LCH risk groups
- Author
-
Hitoshi Takizawa, M. John Hicks, Carl E. Allen, Harshal Abhyankar, Juliana Idoyaga, Stephen J. Simko, Albert Ruzo, Philip J. Lupo, Miriam Merad, Björn E. Clausen, Albert Shih, Marie-Luise Berres, Tricia L. Peters, Rikhia Chakraborty, Kenneth Heym, Hélène Salmon, Sergio A. Lira, Matthew Collin, Marylene Leboeuf, Markus G. Manz, Kenneth L. McClain, Monique Beltrão, Karen Phaik Har Lim, Venetia Bigley, Jeremy Price, University of Zurich, and Merad, M
- Subjects
Male ,Pathology ,endocrine system diseases ,Cellular differentiation ,CD34 ,Antigens, CD34 ,Mice ,0302 clinical medicine ,Langerhans cell histiocytosis ,Bone Marrow ,Risk Factors ,Immunology and Allergy ,skin and connective tissue diseases ,Child ,0303 health sciences ,Cell Differentiation ,3. Good health ,Histiocytosis ,medicine.anatomical_structure ,Phenotype ,Treatment Outcome ,030220 oncology & carcinogenesis ,Child, Preschool ,Antigens, Surface ,2723 Immunology and Allergy ,Female ,Proto-Oncogene Proteins B-raf ,medicine.medical_specialty ,Immunology ,CD11c ,610 Medicine & health ,Biology ,Article ,03 medical and health sciences ,Germline mutation ,medicine ,Animals ,Humans ,Cell Lineage ,Genetic Predisposition to Disease ,Lectins, C-Type ,Progenitor cell ,neoplasms ,030304 developmental biology ,2403 Immunology ,Histocompatibility Antigens Class II ,Infant ,Correction ,Dendritic Cells ,medicine.disease ,Hematopoietic Stem Cells ,digestive system diseases ,CD11c Antigen ,enzymes and coenzymes (carbohydrates) ,Histiocytosis, Langerhans-Cell ,Mannose-Binding Lectins ,10032 Clinic for Oncology and Hematology ,Mutation ,Bone marrow - Abstract
BRAF-V600E expression is identified in hematopoietic progenitor and precursor myeloid dendritic cells in patients with high-risk LCH, and enforced expression of BRAF-V600E in CD11c+ cells recapitulates a high-risk LCH-like phenotype in mice., Langerhans cell histiocytosis (LCH) is a clonal disorder with elusive etiology, characterized by the accumulation of CD207+ dendritic cells (DCs) in inflammatory lesions. Recurrent BRAF-V600E mutations have been reported in LCH. In this study, lesions from 100 patients were genotyped, and 64% carried the BRAF-V600E mutation within infiltrating CD207+ DCs. BRAF-V600E expression in tissue DCs did not define specific clinical risk groups but was associated with increased risk of recurrence. Strikingly, we found that patients with active, high-risk LCH also carried BRAF-V600E in circulating CD11c+ and CD14+ fractions and in bone marrow (BM) CD34+ hematopoietic cell progenitors, whereas the mutation was restricted to lesional CD207+ DC in low-risk LCH patients. Importantly, BRAF-V600E expression in DCs was sufficient to drive LCH-like disease in mice. Consistent with our findings in humans, expression of BRAF-V600E in BM DC progenitors recapitulated many features of the human high-risk LCH, whereas BRAF-V600E expression in differentiated DCs more closely resembled low-risk LCH. We therefore propose classification of LCH as a myeloid neoplasia and hypothesize that high-risk LCH arises from somatic mutation of a hematopoietic progenitor, whereas low-risk disease arises from somatic mutation of tissue-restricted precursor DCs.
- Published
- 2014
21. Targeting Leishmania major Antigens to Dendritic Cells In Vivo Induces Protective Immunity
- Author
-
Ashira Lubkin, Ines Matos, Juliana Idoyaga, Olga Mizenina, and Ralph M. Steinman
- Subjects
0303 health sciences ,Protective immunity ,Multidisciplinary ,business.industry ,Science ,Library science ,Correction ,3. Good health ,03 medical and health sciences ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Medicine ,Translational science ,business ,030304 developmental biology - Abstract
Multiple funding organizations and grants were incorrectly omitted from the Funding Statement. The Funding Statement should read: "Funding was provided by NIH/NIAID grant AI13013 (to R.M.S.), The Rockefeller University Center for Clinical and Translational Science Grant Award Number ULTR000043 (to J.I.), NIH/NIAMS grant 1K99AR062595 (to J.I.), and by the Fundacao para a Ciencia e Tecnologia PhD scholarship SFRH/BD/41073/2007 (to I.M.). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript."
- Published
- 2013
22. Zinc finger transcription factor zDC is a negative regulator required to prevent activation of classical dendritic cells in the steady state
- Author
-
Israel Tojal da Silva, Rafael Casellas, Arito Yamane, Michel C. Nussenzweig, Juliana Idoyaga, Suzannah J. Rihn, Thiago Yukio Kikuchi Oliveira, Alice O. Kamphorst, Matthew M. Meredith, Kai-Hui Yao, Dimitris Skokos, Pierre Guermonprez, and Kang Liu
- Subjects
Chromatin Immunoprecipitation ,Immunology ,Antigen presentation ,chemical and pharmacologic phenomena ,Major histocompatibility complex ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immunology and Allergy ,Animals ,Lymphangiogenesis ,Promoter Regions, Genetic ,Transcription factor ,030304 developmental biology ,Zinc finger transcription factor ,Zinc finger ,Regulation of gene expression ,Mice, Knockout ,0303 health sciences ,biology ,Toll-Like Receptors ,Dendritic Cells ,respiratory system ,Molecular biology ,3. Good health ,Gene Expression Regulation ,biology.protein ,Chromatin immunoprecipitation ,CD8 ,030215 immunology ,Transcription Factors - Abstract
Conventional DCs from mice lacking zDC (also known as Zbtb46) express more MHCII and produce more VEGF in the steady state., Classical dendritic cells (cDCs) process and present antigens to T cells. Under steady-state conditions, antigen presentation by cDCs induces tolerance. In contrast, during infection or inflammation, cDCs become activated, express higher levels of cell surface MHC molecules, and induce strong adaptive immune responses. We recently identified a cDC-restricted zinc finger transcription factor, zDC (also known as Zbtb46 or Btbd4), that is not expressed by other immune cell populations, including plasmacytoid DCs, monocytes, or macrophages. We define the zDC consensus DNA binding motif and the genes regulated by zDC using chromatin immunoprecipitation and deep sequencing. By deleting zDC from the mouse genome, we show that zDC is primarily a negative regulator of cDC gene expression. zDC deficiency alters the cDC subset composition in the spleen in favor of CD8+ DCs, up-regulates activation pathways in steady-state cDCs, including elevated MHC II expression, and enhances cDC production of vascular endothelial growth factor leading to increased vascularization of skin-draining lymph nodes. Consistent with these observations, zDC protein expression is rapidly down-regulated after TLR stimulation. Thus, zDC is a TLR-responsive, cDC-specific transcriptional repressor that is in part responsible for preventing cDC maturation in the steady state.
- Published
- 2012
23. Skin Langerin + Dendritic Cells Transport Intradermally Injected Anti–DEC-205 Antibodies but Are Not Essential for Subsequent Cytotoxic CD8 + T Cell Responses
- Author
-
Adrien Kissenpfennig, Patrizia Stoitzner, Bernhard Haid, Bernard Malissen, Christoph H. Tripp, Nikolaus Romani, Vincent Flacher, Juliana Idoyaga, and Innsbruck Medical University [Austria] (IMU)
- Subjects
Langerin ,T cell ,Immunology ,chemical and pharmacologic phenomena ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Antigen ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,ComputingMilieux_MISCELLANEOUS ,030304 developmental biology ,0303 health sciences ,integumentary system ,biology ,hemic and immune systems ,TLR7 ,3. Good health ,medicine.anatomical_structure ,Lymphatic system ,biology.protein ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,CD8 ,030215 immunology - Abstract
Incorporation of Ags by dendritic cells (DCs) increases when Ags are targeted to endocytic receptors by mAbs. We have previously demonstrated in the mouse that mAbs against C-type lectins administered intradermally are taken up by epidermal Langerhans cells (LCs), dermal Langerinneg DCs, and dermal Langerin+ DCs in situ. However, the relative contribution of these skin DC subsets to the induction of immune responses after Ag targeting has not been addressed in vivo. We show in this study that murine epidermal LCs and dermal DCs transport intradermally injected mAbs against the lectin receptor DEC-205/CD205 in vivo. Skin DCs targeted in situ with mAbs migrated through lymphatic vessels in steady state and inflammation. In the skin-draining lymph nodes, targeting mAbs were found in resident CD8α+ DCs and in migrating skin DCs. More than 70% of targeted DCs expressed Langerin, including dermal Langerin+ DCs and LCs. Numbers of targeted skin DCs in the nodes increased 2-3-fold when skin was topically inflamed by the TLR7 agonist imiquimod. Complete removal of the site where OVA-coupled anti–DEC-205 had been injected decreased endogenous cytotoxic responses against OVA peptide-loaded target cells by 40–50%. Surprisingly, selective ablation of all Langerin+ skin DCs in Langerin-DTR knock-in mice did not affect such responses independently of the adjuvant chosen. Thus, in cutaneous immunization strategies where Ag is targeted to DCs, Langerin+ skin DCs play a major role in transport of anti–DEC-205 mAb, although Langerinneg dermal DCs and CD8α+ DCs are sufficient to subsequent CD8+ T cell responses.
- Published
- 2012
- Full Text
- View/download PDF
24. Microbial stimulation fully differentiates monocytes to DC-SIGN/CD209(+) dendritic cells for immune T cell areas
- Author
-
Anthony Rodriguez, M. Paula Longhi, Chae Gyu Park, Ralph M. Steinman, Hyein Koh, Durga Bhavani Dandamudi, Kate L. Jeffrey, Courtney Kluger, Elina Shrestha, Jae-Hoon Choi, Godwin Nchinda, Cheolho Cheong, Juliana Idoyaga, Klara Velinzon, Maggi Pack, Robert M. Anthony, and Ines Matos
- Subjects
Receptors, CCR7 ,T cell ,CD14 ,T-Lymphocytes ,Antigen presentation ,HUMDISEASE ,Lipopolysaccharide Receptors ,chemical and pharmacologic phenomena ,Receptors, Cell Surface ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Monocytes ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,medicine ,Escherichia coli ,Animals ,Lectins, C-Type ,L-Selectin ,030304 developmental biology ,0303 health sciences ,Antigen Presentation ,Mice, Inbred BALB C ,Follicular dendritic cells ,Biochemistry, Genetics and Molecular Biology(all) ,Monocyte ,hemic and immune systems ,Cell Differentiation ,Dendritic cell ,Dendritic Cells ,DC-SIGN ,Mice, Inbred C57BL ,Toll-Like Receptor 4 ,medicine.anatomical_structure ,CELLIMMUNO ,Immunology ,biology.protein ,CELLBIO ,Lymph Nodes ,Cell Adhesion Molecules ,030215 immunology - Abstract
SummaryDendritic cells (DCs), critical antigen-presenting cells for immune control, normally derive from bone marrow precursors distinct from monocytes. It is not yet established if the large reservoir of monocytes can develop into cells with critical features of DCs in vivo. We now show that fully differentiated monocyte-derived DCs (Mo-DCs) develop in mice and DC-SIGN/CD209a marks the cells. Mo-DCs are recruited from blood monocytes into lymph nodes by lipopolysaccharide and live or dead gram-negative bacteria. Mobilization requires TLR4 and its CD14 coreceptor and Trif. When tested for antigen-presenting function, Mo-DCs are as active as classical DCs, including cross-presentation of proteins and live gram-negative bacteria on MHC I in vivo. Fully differentiated Mo-DCs acquire DC morphology and localize to T cell areas via L-selectin and CCR7. Thus the blood monocyte reservoir becomes the dominant presenting cell in response to select microbes, yielding DC-SIGN+ cells with critical functions of DCs.
- Published
- 2010
25. Dendritic cells require a systemic type I interferon response to mature and induce CD4+ Th1 immunity with poly IC as adjuvant
- Author
-
Marina Caskey, Ralph M. Steinman, Andres M. Salazar, M. Paula Longhi, Courtney Kluger, Christine Trumpfheller, Ines Matos, Juliana Idoyaga, and Marco Colonna
- Subjects
CD4-Positive T-Lymphocytes ,T cell ,Immunology ,Antigen presentation ,Bone Marrow Cells ,Mice, Inbred Strains ,Receptor, Interferon alpha-beta ,Biology ,Lymphocyte Activation ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Adjuvants, Immunologic ,Interferon ,T-Lymphocyte Subsets ,medicine ,Immunology and Allergy ,Animals ,030304 developmental biology ,Mice, Knockout ,0303 health sciences ,Chimera ,Dendritic cell ,Dendritic Cells ,Th1 Cells ,Acquired immune system ,Interleukin-12 ,Immunity, Innate ,3. Good health ,medicine.anatomical_structure ,Poly I-C ,TLR3 ,Interferon Type I ,Interleukin 12 ,Cytokines ,Interferon type I ,Spleen ,030215 immunology ,medicine.drug ,Signal Transduction - Abstract
Relative to several other toll-like receptor (TLR) agonists, we found polyinosinic:polycytidylic acid (poly IC) to be the most effective adjuvant for Th1 CD4+ T cell responses to a dendritic cell (DC)–targeted HIV gag protein vaccine in mice. To identify mechanisms for adjuvant action in the intact animal and the polyclonal T cell repertoire, we found poly IC to be the most effective inducer of type I interferon (IFN), which was produced by DEC-205+ DCs, monocytes, and stromal cells. Antibody blocking or deletion of type I IFN receptor showed that IFN was essential for DC maturation and development of CD4+ immunity. The IFN-AR receptor was directly required for DCs to respond to poly IC. STAT 1 was also essential, in keeping with the type I IFN requirement, but not type II IFN or IL-12 p40. Induction of type I IFN was mda5 dependent, but DCs additionally used TLR3. In bone marrow chimeras, radioresistant and, likely, nonhematopoietic cells were the main source of IFN, but mda5 was required in both marrow–derived and radioresistant host cells for adaptive responses. Therefore, the adjuvant action of poly IC requires a widespread innate type I IFN response that directly links antigen presentation by DCs to adaptive immunity.
- Published
- 2009
26. Epidermal Langerhans Cells Rapidly Capture and Present Antigens from C-Type Lectin-Targeting Antibodies Deposited in the Dermis
- Author
-
Bernhard Haid, Barbara Del Frari, Patrizia Stoitzner, Vincent Flacher, Chae Gyu Park, Franz Koch, Juliana Idoyaga, Susanne Ebner, Ralph M. Steinman, Christoph H. Tripp, Nikolaus Romani, and Innsbruck Medical University [Austria] (IMU)
- Subjects
Langerhans cell ,Injections, Intradermal ,Langerin ,T-Lymphocytes ,Antigen presentation ,Human skin ,chemical and pharmacologic phenomena ,Dermatology ,Biochemistry ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Dermis ,Antigen ,Cell Movement ,C-type lectin ,medicine ,Animals ,Humans ,Lectins, C-Type ,Molecular Biology ,Cells, Cultured ,ComputingMilieux_MISCELLANEOUS ,030304 developmental biology ,Antigen Presentation ,Mice, Inbred BALB C ,0303 health sciences ,biology ,integumentary system ,Antibodies, Monoclonal ,hemic and immune systems ,Dendritic cell ,Cell Biology ,3. Good health ,Cell biology ,Mice, Inbred C57BL ,Mannose-Binding Lectins ,medicine.anatomical_structure ,Epidermal Cells ,Langerhans Cells ,Antigens, Surface ,Immunology ,biology.protein ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,Epidermis ,Cell Division ,030215 immunology - Abstract
Antigen-presenting cells can capture antigens that are deposited in the skin, including vaccines given subcutaneously. These include different dendritic cells (DCs) such as epidermal Langerhans cells (LCs), dermal DCs, and dermal langerin + DCs. To evaluate access of dermal antigens to skin DCs, we used mAb to two C-type lectin endocytic receptors, DEC-205/CD205 and langerin/CD207. When applied to murine and human skin explant cultures, these mAbs were efficiently taken up by epidermal LCs. In addition, anti-DEC-205 targeted langerin + CD103 + and langerin − CD103 − mouse dermal DCs. Unexpectedly, intradermal injection of either mAb, but not isotype control, resulted in strong and rapid labeling of LCs in situ , implying that large molecules can diffuse through the basement membrane into the epidermis. Epidermal LCs targeted in vivo by ovalbumin-coupled anti-DEC-205 potently presented antigen to CD4 + and CD8 + T cells in vitro . However, to our surprise, LCs targeted through langerin were unable to trigger T-cell proliferation. Thus, epidermal LCs have a major function in uptake of lectin-binding antibodies under standard vaccination conditions.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.