Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Cell ; 169(4): 750-765.e17, 2017 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-28475900

RESUMO

To guide the design of immunotherapy strategies for patients with early stage lung tumors, we developed a multiscale immune profiling strategy to map the immune landscape of early lung adenocarcinoma lesions to search for tumor-driven immune changes. Utilizing a barcoding method that allows a simultaneous single-cell analysis of the tumor, non-involved lung, and blood cells, we provide a detailed immune cell atlas of early lung tumors. We show that stage I lung adenocarcinoma lesions already harbor significantly altered T cell and NK cell compartments. Moreover, we identified changes in tumor-infiltrating myeloid cell (TIM) subsets that likely compromise anti-tumor T cell immunity. Paired single-cell analyses thus offer valuable knowledge of tumor-driven immune changes, providing a powerful tool for the rational design of immune therapies. VIDEO ABSTRACT.


Assuntos
Adenocarcinoma/imunologia , Adenocarcinoma/patologia , Imunidade Inata , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Análise de Célula Única/métodos , Adenocarcinoma de Pulmão , Células Dendríticas/patologia , Humanos , Células Matadoras Naturais/patologia , Macrófagos/patologia , Linfócitos T/patologia , Microambiente Tumoral
2.
BMC Cancer ; 23(1): 129, 2023 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-36755259

RESUMO

BACKGROUND: The tumor microenvironment (TME) in breast cancer plays a vital role in occurrence, development, and therapeutic responses. However, immune and stroma constituents in the TME are major obstacles to understanding and treating breast cancer. We evaluated the significance of TME-related genes in breast cancer. METHODS: Invasive breast cancer (BRCA) samples were retrieved from the TCGA and GEO databases. Stroma and immune scores of samples as well as the proportion of tumor infiltrating immune cells (TICs) were calculated using the ESTIMATE and CIBERSORT algorithms. TME-related differentially expressed genes (DEGs) were analyzed by a protein interaction (PPI) network and univariate Cox regression to determine CD1C as a hub gene. Subsequently, the prognostic value of CD1C, its response to immunotherapy, and its mechanism in the TME were further studied. RESULTS: In BRCA, DEGs were determined to identify CD1C as a hub gene. The expression level of CD1C in BRCA patients was verified based on the TCGA database, polymerase chain reaction (PCR) results, and western blot analysis. Immunohistochemical staining (IHC) results revealed a correlation between prognosis, clinical features, and CD1C expression in BRCA. Enrichment analysis of GSEA and GSVA showed that CD1C participates in immune-associated signaling pathways. CIBERSORT showed that CD1C levels were associated with tumor immune infiltrating cells (TILs), such as different kinds of T cells. Gene co-expression analysis showed that CD1C and the majority of immune-associated genes were co-expressed in BRCA. In renal cell carcinoma, patients with a high expression of CD1C had a better immunotherapy effect. CONCLUSION: CD1C is an important part of the TME and participates in immune activity regulation in breast tumors. CD1C is expected to become a prognostic marker and a new treatment target for breast cancer.


Assuntos
Antígenos CD1 , Neoplasias da Mama , Glicoproteínas , Feminino , Humanos , Antígenos CD1/genética , Mama , Neoplasias da Mama/genética , Glicoproteínas/genética , Prognóstico , Microambiente Tumoral/genética
3.
J Biol Chem ; 297(4): 101197, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34536421

RESUMO

Whereas proteolytic cleavage is crucial for peptide presentation by classical major histocompatibility complex (MHC) proteins to T cells, glycolipids presented by CD1 molecules are typically presented in an unmodified form. However, the mycobacterial lipid antigen mannosyl-ß1-phosphomycoketide (MPM) may be processed through hydrolysis in antigen presenting cells, forming mannose and phosphomycoketide (PM). To further test the hypothesis that some lipid antigens are processed, and to generate antigens that lead to defined epitopes for future tuberculosis vaccines or diagnostic tests, we aimed to create hydrolysis-resistant MPM variants that retain their antigenicity. Here, we designed and tested three different, versatile synthetic strategies to chemically stabilize MPM analogs. Crystallographic studies of CD1c complexes with these three new MPM analogs showed anchoring of the lipid tail and phosphate group that is highly comparable to nature-identical MPM, with considerable conformational flexibility for the mannose head group. MPM-3, a difluoromethylene-modified version of MPM that is resistant to hydrolysis, showed altered recognition by cells, but not by CD1c proteins, supporting the cellular antigen processing hypothesis. Furthermore, the synthetic analogs elicited T cell responses that were cross-reactive with nature-identical MPM, fulfilling important requirements for future clinical use.


Assuntos
Antígenos de Bactérias/química , Antígenos CD1/química , Glicolipídeos/química , Glicoproteínas/química , Mycobacterium tuberculosis/química , Fosfolipídeos/química , Linfócitos T/química , Antígenos de Bactérias/imunologia , Antígenos CD1/imunologia , Linhagem Celular Transformada , Cristalografia por Raios X , Glicolipídeos/imunologia , Glicoproteínas/imunologia , Humanos , Mycobacterium tuberculosis/imunologia , Fosfolipídeos/imunologia , Linfócitos T/imunologia
4.
FASEB J ; 34(7): 9269-9284, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32413173

RESUMO

Monocytic cells perform crucial homeostatic and defensive functions. However, their fate and characterization at the transcriptomic level in human tissues are partially understood, often as a consequence of the lack of specific markers allowing their unequivocal identification. The 6-sulfo LacNAc (slan) antigen identifies a subset of non-classical (NC) monocytes in the bloodstream, namely the slan+ -monocytes. In recent studies, we and other groups have reported that, in tonsils, slan marks dendritic cell (DC)-like cells, as defined by morphological, phenotypical, and functional criteria. However, subsequent investigations in lymphomas have uncovered a significant heterogeneity of tumor-infiltrating slan+ -cells, including a macrophage-like state. Based on their emerging role in tissue inflammation and cancer, herein we investigated slan+ -cell fate in tonsils by using a molecular-based approach. Hence, RNA from tonsil slan+ -cells, conventional CD1c+ DCs (cDC2) and CD11b+ CD14+ -macrophages was subjected to gene expression analysis. For comparison, transcriptomes were also obtained from blood cDC2, classical (CL), intermediate (INT), NC, and slan+ -monocytes. Data demonstrate that the main trajectory of human slan+ -monocytes infiltrating the tonsil tissue is toward a macrophage-like population, displaying molecular features distinct from those of tonsil CD11b+ CD14+ -macrophages and cDC2. These findings provide a novel view on the terminal differentiation path of slan+ -monocytes, which is relevant for inflammatory diseases and lymphomas.


Assuntos
Amino Açúcares/metabolismo , Células Dendríticas/metabolismo , Macrófagos/metabolismo , Monócitos/metabolismo , Tonsila Palatina/metabolismo , Tonsilite/genética , Estudos de Casos e Controles , Células Cultivadas , Células Dendríticas/citologia , Perfilação da Expressão Gênica , Humanos , Macrófagos/citologia , Monócitos/citologia , Tonsila Palatina/citologia , Tonsilite/metabolismo , Tonsilite/patologia
5.
J Infect Dis ; 221(2): 201-213, 2020 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-31647546

RESUMO

BACKGROUND: For the purpose of studying functional human dendritic cells (DCs) in a humanized mouse model that mimics the human immune system (HIS), a model referred to as HIS mice was established. METHODS: Human immune system mice were made by engrafting NOD/SCID/IL2Rgammanull (NSG) mice with human hematopoietic stem cells (HSCs) following the transduction of genes encoding human cytokines and human leukocyte antigen (HLA)-A2.1 by adeno-associated virus serotype 9 (AAV9) vectors. RESULTS: Our results indicate that human DC subsets, such as CD141+CD11c+ and CD1c+CD11c+ myeloid DCs, distribute throughout several organs in HIS mice including blood, bone marrow, spleen, and draining lymph nodes. The CD141+CD11c+ and CD1c+CD11c+ human DCs isolated from HIS mice immunized with adenoviruses expressing malaria/human immunodeficiency virus (HIV) epitopes were able to induce the proliferation of malaria/HIV epitopes-specific human CD8+ T cells in vitro. Upregulation of CD1c was also observed in human CD141+ DCs 1 day after immunization with the adenovirus-based vaccines. CONCLUSIONS: Establishment of such a humanized mouse model that mounts functional human DCs enables preclinical assessment of the immunogenicity of human vaccines in vivo.


Assuntos
Vacinas contra Adenovirus/imunologia , Antígenos de Superfície/imunologia , Células Dendríticas/imunologia , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Trombomodulina
6.
Immunol Invest ; 48(7): 704-718, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31044626

RESUMO

Immunoglobulin-like transcript (ILT) 4 is an inhibitory immune receptor of the immunoglobulin superfamily, which could deliver inhibitory signals and induce immunosuppression. The significance of the expression of ILT4 in mDCs subsets in patients with hepatocellular carcinoma (HCC) remains unclear. In this study, the frequency of mDCs subsets in the peripheral blood of 121 patients with HCC and 103 normal controls, and in the tumor and tumor free liver tissues (TFL) of 43 HCC patients was analyzed by flow cytometry. Then, the expressions of ILT4 in mDCs subsets in the microenvironment of liver cancer were also analyzed. Results showed that the percentage of CD1c+ subset was dramatically decreased in peripheral blood mononuclear cells (PBMCs) of HCC patients compared with normal controls, and also significantly decreased in tumor tissue compared with the TFL. The decreased of CD1c+ subset in blood could be a diagnostic factor for HCC with the area under the receiver operating characteristic curve 0.975 (P < 0.01). The percentage of ILT4+CD1c+ subset was dramatically increased in tumor than that of TFL and blood. There were significant correlations between the percentage of ILT4+ in CD1c+ subset in tumor and that of in blood. The percentage of ILT4+CD1c+ subset in tumor tissue was strongly associated with the Edmondson-Steiner stage in HCC (P = 0.03). Furthermore, the capacity of ILT4+CD1c+ subset producing IFN-γ was lower than ILT4- CD1c subset in PBMC of HCC patients following Poly I:C stimulation. Taken together, the increased ILT4+CD1c+ subset in tumor tissue might play an important role in immune suppression for patients with HCC.


Assuntos
Carcinoma Hepatocelular/imunologia , Células Dendríticas/imunologia , Neoplasias Hepáticas/imunologia , Glicoproteínas de Membrana/imunologia , Células Mieloides/imunologia , Receptores Imunológicos/imunologia , Antígenos CD1/imunologia , Biomarcadores Tumorais/imunologia , Carcinoma Hepatocelular/sangue , Carcinoma Hepatocelular/patologia , Células Dendríticas/patologia , Feminino , Glicoproteínas/imunologia , Humanos , Interferon gama/metabolismo , Neoplasias Hepáticas/sangue , Neoplasias Hepáticas/patologia , Masculino , Pessoa de Meia-Idade , Células Mieloides/patologia , Poli I-C/farmacologia , Curva ROC , Microambiente Tumoral
7.
Immunol Cell Biol ; 96(4): 390-400, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29344995

RESUMO

Mice reconstituted with human hematopoietic stem cells are valuable models to study aspects of the human immune system in vivo. We describe a humanized mouse model (hu mice) in which fully functional human CD141+ and CD1c+ myeloid and CD123+ plasmacytoid dendritic cells (DC) develop from human cord blood CD34+ cells in immunodeficient mice. CD141+ DC are the human equivalents of murine CD8+ /CD103+ DC which are essential for the induction of tumor-inhibitory cytotoxic T lymphocyte responses, making them attractive targets to exploit for the development of new cancer immunotherapies. We used CD34+ -engrafted NSG-A2 mice to investigate activation of DC subsets by synthetic dsRNA or ssRNA analogs polyinosinic-polycytidylic acid/poly I:C and Resiquimod/R848, agonists for TLR3 and TLR8, respectively, both of which are expressed by CD141+ DC. Injection of hu mice with these agonists resulted in upregulation of costimulatory molecules CD80, CD83 and CD86 by CD141+ and CD1c+ DC alike, and their combination further enhanced expression of these molecules by both subsets. When combined, poly I:C and R848 enhanced serum levels of key cytokines associated with cross-presentation and the induction of cytotoxic T lymphocyte responses including IFN-α, IFN-ß, IL-12 and CXCL10. These data advocate a combination of poly I:C and R848 TLR agonists as means of activating human DC for immunotherapy.


Assuntos
Antígenos CD1/metabolismo , Antígenos de Superfície/metabolismo , Células Dendríticas/imunologia , Receptores Toll-Like/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Citocinas/sangue , Células Dendríticas/efeitos dos fármacos , Humanos , Imidazóis/farmacologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Tecido Linfoide/efeitos dos fármacos , Tecido Linfoide/metabolismo , Camundongos , Poli I-C/farmacologia , Receptores Toll-Like/agonistas , Regulação para Cima/efeitos dos fármacos
8.
Cell Mol Life Sci ; 72(22): 4309-25, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26243730

RESUMO

The method of choice for the development of new vaccines is to target distinct dendritic cell subsets with antigen in vivo and to harness their function in situ to enhance cell-mediated immunity or induce tolerance to specific antigens. The innate functions of dendritic cells themselves may also be targeted by inhibitors or activators that would target a specific function such as interferon production, potentially important in autoimmune disease and chronic viral infections. Importantly targeting dendritic cells requires detailed knowledge of both the surface phenotype and function of each dendritic cell subset, including how they may respond to different types of vaccine adjuvants, their ability to produce soluble mediators and to process and present antigens and induce priming of naïve T cells. This review summarizes our knowledge of the functional attributes of the human dendritic cell subsets in the steady state and upon activation and their roles in human disease.


Assuntos
Apresentação de Antígeno/imunologia , Células Dendríticas/imunologia , Doenças do Sistema Imunitário/imunologia , Imunidade Celular/imunologia , Linfócitos T/imunologia , Antígenos CD1/imunologia , Antígenos CD1/metabolismo , Citocinas/imunologia , Citocinas/metabolismo , Células Dendríticas/metabolismo , Glicoproteínas/imunologia , Glicoproteínas/metabolismo , Humanos , Infecções/imunologia , Ativação Linfocitária/imunologia
9.
Clin Immunol ; 158(1): 35-46, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25784146

RESUMO

In this study we investigated the role of blood CD1c(+) myeloid dendritic cells 1 (mDC1), a key mDC subtype, in patients with autoimmune uveitis. We observed a significant increase of blood CD1c(+) mDC1 in uveitis patients. The increased CD1c(+) mDC1 exhibited high HLADR expression and less antigen uptake. CD1c(+) mDC1 were divided into two subpopulations. CD1c(hi) mDC1 subpopulation showed less antigen uptake and higher HLADR expression compared to CD1c(lo) mDC1 subpopulation. Importantly, the CD1c(hi) mDC1 subpopulation was increased in uveitis patients. In vitro, mature monocyte-derived dendritic cells (MoDCs), characterized by lower levels of antigen uptake, induced more CD4(+)CD62L(-) T helper cell proliferation. The mature phenotype and function of CD1c(+) mDC1 were regulated by TNFα via a p38 MAPK-dependent pathway. These data show that alterations in the systemic immune response are involved in the pathogenesis of autoimmune uveitis and invite the therapeutic possibility of attenuating uveitis by manipulating blood CD1c(+) mDC1.


Assuntos
Antígenos CD1/imunologia , Doenças Autoimunes/imunologia , Células Dendríticas/imunologia , Glicoproteínas/imunologia , Fator de Necrose Tumoral alfa/imunologia , Uveíte/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/imunologia , Adolescente , Adulto , Idoso , Antígenos CD1/metabolismo , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Feminino , Glicoproteínas/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Células Mieloides/citologia , Transdução de Sinais , Linfócitos T Auxiliares-Indutores/imunologia , Adulto Jovem
10.
Artigo em Inglês | MEDLINE | ID: mdl-38907868

RESUMO

Lung adenocarcinoma (LUAD) is the most frequent type of lung cancer with a high mortality rate. Here, we aim to explore novel immune-related biomarkers for LUAD patients. Datasets, mRNA expression profiles, and clinical data concerned with LUAD were obtained from Gene Expression Omnibus (GEO) database and The Cancer Genome Atlas (TCGA), respectively. Differential expression analysis was performed to obtain differentially expressed genes (DEGs). Based on DEGs, we conducted functional enrichment analyses. Subsequently, Kaplan­Meier (KM) was performed to analyze survival differences among different groups. Furthermore, immune cell infiltration proportion was calculated by CIBERSORT and TIMER. The relationship between gene and immune response was analyzed using Tumor Immune System Interactions (TISIDB) database. Finally, Pearson correlation analysis was performed between CD1C and six immune checkpoints. We identified dendritic cells (DCs)-related expression profiles from four LUAD samples. DCs' immune marker CD1C in LUAD was selected by univariate Cox regression analysis. Low CD1C expression patients had a poor prognosis. A total of 332 DEGs were identified in high and low CD1C expression groups, which primarily enriched in 348 GO terms and 30 KEGG pathways. There were significant differences in the infiltration proportion of 17 immune cells between high and low CD1C expression groups. Most immunomodulators, chemokines, and chemokine receptors were positively associated with CD1C expression. Six immune checkpoints were also positively correlated with CD1C expression. DCs related immunomarker CD1C probably plays a pivotal part in prognosis and immunotherapy of LUAD via a joint analysis of single-cell and bulk sequencing data.

11.
Elife ; 122023 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-37042831

RESUMO

Background: Type I interferons (IFNs) promote the expansion of subsets of CD1c+ conventional dendritic cells (CD1c+ DCs), but the molecular basis of CD1c+ DCs involvement in conditions not associated without elevated type I IFNs remains unclear. Methods: We analyzed CD1c+ DCs from two cohorts of non-infectious uveitis patients and healthy donors using RNA-sequencing followed by high-dimensional flow cytometry to characterize the CD1c+ DC populations. Results: We report that the CD1c+ DCs pool from patients with non-infectious uveitis is skewed toward a gene module with the chemokine receptor CX3CR1 as the key hub gene. We confirmed these results in an independent case-control cohort and show that the disease-associated gene module is not mediated by type I IFNs. An analysis of peripheral blood using flow cytometry revealed that CX3CR1+ DC3s were diminished, whereas CX3CR1- DC3s were not. Stimulated CX3CR1+ DC3s secrete high levels of inflammatory cytokines, including TNF-alpha, and CX3CR1+ DC3 like cells can be detected in inflamed eyes of patients. Conclusions: These results show that CX3CR1+ DC3s are implicated in non-infectious uveitis and can secrete proinflammatory mediators implicated in its pathophysiology. Funding: The presented work is supported by UitZicht (project number #2014-4, #2019-10, and #2021-4). The funders had no role in the design, execution, interpretation, or writing of the study.


Assuntos
Transcriptoma , Uveíte , Humanos , Antígenos CD1/análise , Citocinas , Células Dendríticas/fisiologia , Uveíte/genética , Citometria de Fluxo , Receptor 1 de Quimiocina CX3C/genética
12.
PeerJ ; 11: e16618, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38099311

RESUMO

Background: Tumor mutational burden (TMB) is a valuable prognostic biomarker. This study explored the predictive value of TMB and the potential association between TMB and immune infiltration in diffuse large B-cell lymphoma (DLBCL). Methods: We downloaded the gene expression profile, somatic mutation, and clinical data of DLBCL patients from The Cancer Genome Atlas (TCGA) database. We classified the samples into high-and low-TMB groups to identify differentially expressed genes (DEGs). Functional enrichment analyses were performed to determine the biological functions of the DEGs. We utilized the cell-type identification by estimating relative subsets of RNA transcripts (CIBERSORT) algorithm to estimate the abundance of 22 immune cells, and the significant difference was determined by the Wilcoxon rank-sum test between the high- and low-TMB group. Hub gene had been screened as the prognostic TMB-related immune biomarker by the combination of the Immunology Database and Analysis Portal (ImmPort) database and the univariate Cox analysis from the Gene Expression Omnibus (GEO) database including six DLBCL datasets. Various database applications such as Tumor Immune Estimation Resource (TIMER), CellMiner, konckTF, and Genotype-Tissue Expression (GTEx) verified the functions of the target gene. Wet assay confirmed the target gene expression at RNA and protein levels in DLBCL tissue and cell samples. Results: Single nucleotide polymorphism (SNP) occurred more frequently than insertion and deletion, and C > T was the most common single nucleotide variant (SNV) in DLBCL. Survival analysis showed that the high-TMB group conferred poor survival outcomes. A total of 62 DEGs were obtained, and 13 TMB-related immune genes were identified. Univariate Cox analysis results illustrated that CD1c mutation was associated with lower TMB and manifested a satisfactory clinical prognosis by analysis of large samples from the GEO database. In addition, infiltration levels of immune cells in the high-TMB group were lower. Using the TIMER database, we systematically analyzed that the expression of CD1c was positively correlated with B cells, neutrophils, and dendritic cells and negatively correlated with CD8+ T cells, CD4+ T cells, and macrophages. Drug sensitivity showed a significant positive correlation between CD1c expression level and clinical drug sensitivity from the CellMiner database. CREB1, AHR, and TOX were used to comprehensively explore the regulation of CD1c-related transcription factors and signaling pathways by the KnockTF database. We searched the GETx database to compare the mRNA expression levels of CD1c between DLBCL and normal tissues, and the results suggested a significant difference between them. Moreover, wet experiments were conducted to verify the high expression of CD1c in DLBCL at the RNA and protein levels. Conclusions: Higher TMB correlated with poor survival outcomes and inhibited the immune infiltrates in DLBCL. Our results suggest that CD1c is a TMB-related prognostic biomarker.


Assuntos
Linfoma Difuso de Grandes Células B , Humanos , Algoritmos , Linfócitos B , Biomarcadores , Linfoma Difuso de Grandes Células B/genética , RNA
13.
Mol Immunol ; 132: 126-131, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33582549

RESUMO

CD1-restricted T cells were first described over 30 years ago along with the cloning of the CD1 family. Around the same time, invariant Natural Killer cells (iNKT) were identified based on invariant TCR-alpha chains with additional expression of natural killer (NK) cell markers. About 5 years later, iNKT were shown to react with CD1d. Since then, iNKT have been shown to be a major population of CD1d-restricted T cells in humans and many animals. Like NK cells, iNKT are innate lymphocytes with rapid and wide-ranging effector potential. These activities include cytotoxicity and an unusually broad and high-level cytokine production. The development of highly-specific methods of isolating, stimulating, expanding or depleting these relatively rare cells and controlling their potent activities has stimulated considerable interest in therapeutic targeting of iNKT cells. Potential applications include cancers, inflammatory, infectious and autoimmune among other diseases. To date, most trials have targeted various cancers, there are 2 published trials in viral hepatitis and one in sickle cell lung disease. Uniform safety, evidence of immunologic activity and increasingly clinical efficacy have been seen. Approaches to targeting iNKT cells in clinical development include highly specific natural glycolipid ligands presented by CD1d and chemical analogues thereof and monoclonal antibody-based targeting of iNKT cells. In the case of iNKT cell-based therapies, novel approaches include arming them with Chimeric Antigen Receptors (CARs) and recombinant TCRs (rTCR), gene editing and allogeneic use. Controlling the iTCR:CD1d molecular interaction and consequences is a unique and promising therapeutic technology.


Assuntos
Antígenos CD1/imunologia , Células T Matadoras Naturais/imunologia , Animais , Humanos , Células Matadoras Naturais/imunologia , Ativação Linfocitária/imunologia , Receptores de Antígenos de Linfócitos T/imunologia
14.
Front Immunol ; 11: 559166, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33101275

RESUMO

Currently three bona fide dendritic cell (DC) types are distinguished in human blood. Herein we focus on type 2 DCs (DC2s) and compare the three defining markers CD1c, CD172, and CD301. When using CD1c to define DC2s, a CD14+ and a CD14- subset can be detected. The CD14+ subset shares features with monocytes, and this includes substantially higher expression levels for CD64, CD115, CD163, and S100A8/9. We review the current knowledge of these CD1c+CD14+ cells as compared to the CD1c+CD14- cells with respect to phenotype, function, transcriptomics, and ontogeny. Here, we discuss informative mutations, which suggest that two populations have different developmental requirements. In addition, we cover subsets of CD11c+CD8- DC2s in the mouse, where CLEC12A+ESAMlow cells, as compared to the CLEC12A-ESAMhigh subset, also express higher levels of monocyte-associated markers CD14, CD3, and CD115. Finally, we summarize, for both man and mouse, the data on lower antigen presentation and higher cytokine production in the monocyte-marker expressing DC2 subset, which demonstrate that the DC2 subsets are also functionally distinct.


Assuntos
Antígenos CD1/metabolismo , Células Dendríticas/imunologia , Glicoproteínas/metabolismo , Monócitos/imunologia , Animais , Antígenos de Diferenciação/metabolismo , Assialoglicoproteínas/metabolismo , Diferenciação Celular , Linhagem da Célula , Citocinas/metabolismo , Humanos , Imunidade Celular , Lectinas Tipo C/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Receptores Imunológicos/metabolismo
15.
Cancers (Basel) ; 11(6)2019 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-31234526

RESUMO

Leukemias are clonal proliferative disorders arising from immature leukocytes in the bone marrow. While the advent of targeted therapies has improved survival in certain subtypes, relapse after initial therapy is a major problem. Dendritic cell (DC) vaccination has the potential to induce tumor-specific T cells providing long-lasting, anti-tumor immunity. This approach has demonstrated safety but limited clinical success until recently, as DC vaccination faces several barriers in both solid and hematological malignancies. Importantly, vaccine-mediated stimulation of protective immune responses is hindered by the aberrant production of immunosuppressive factors by cancer cells which impede both DC and T cell function. Leukemias present the additional challenge of severely disrupted hematopoiesis owing to both cytogenic defects in hematopoietic progenitors and an abnormal hematopoietic stem cell niche in the bone marrow; these factors accentuate systemic immunosuppression and DC malfunction. Despite these obstacles, several recent clinical trials have caused great excitement by extending survival in Acute Myeloid Leukemia (AML) patients through DC vaccination. Here, we review the phenotype and functional capacity of DCs in leukemia and approaches to harness DCs in leukemia patients. We describe the recent clinical successes in AML and detail the multiple new strategies that might enhance prognosis in AML and other leukemias.

16.
Front Immunol ; 10: 2829, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31921114

RESUMO

Advanced non-small cell lung cancer (NSCLC) leads to a high death rate in patients and is a major threat to human health. NSCLC induces an immune suppressive microenvironment and escapes from immune surveillance in vivo. At present, the molecular mechanisms of NSCLC immunopathogenesis and the immune suppressive microenvironment induced by NSCLC have not been fully elucidated. Here, we focus on the effect of NSCLC cells on the development and differentiation of human CD1c+ conventional dendritic cell (DC) subsets mediated by CD205 and CD103. The peripheral blood mononuclear cells (PBMCs) were isolated from NSCLC patients and healthy donors. DCs were induced and cocultured with primary NSCLC cells or tumor cell line H1299. DCs without incubation with tumor cells are control. The protein expression of costimulatory molecules such as CD80 and CD86, HLA-DR, pro-/anti-inflammatory cytokines such as IL-10 and IL-12, and CD205 and CD103 on CD1c+ DCs was detected by flow cytometry. Our data revealed two new subpopulations of human CD1c+ DCs (CD1c+CD205+CD103+ and CD1c+CD205+CD103- DC) in healthy donors and NSCLC patients. NSCLC cells modulate the development of the CD1c+CD205+CD103+ DC and CD1c+CD205+CD103- DC subpopulations in vitro and ex vivo. NSCLC cells also suppress the expression of signal molecules such as CD40, CD80, CD86, and HLA-DR on CD1c+ DCs. In addition, the production of pro-inflammatory cytokines, including IL-12 and IL-23, is downregulated by NSCLC cells; however, the secretion of anti-inflammatory cytokines, such as IL-10 and IL-27, by CD1c+ DCs is upregulated by NSCLC cells. Our results suggest that NSCLC cells may induce immune tolerogenic DCs, which block DC-mediated anti-tumor immunity in NSCLC patients. Our data may be helpful in revealing new cellular mechanisms related to the induction of tolerogenic CD1c+ DCs by NSCLCs and the development of an immune suppressive microenvironment that causes tumor cells to escape immune surveillance. Our results indicate a potential role for CD1c+ DC subsets mediated by CD205 and CD103 in DC-mediated immunotherapy to target NSCLC in the future.


Assuntos
Antígenos CD1/metabolismo , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Células Dendríticas/metabolismo , Glicoproteínas/metabolismo , Neoplasias Pulmonares/metabolismo , Antígenos CD/metabolismo , Carcinoma Pulmonar de Células não Pequenas/etiologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/terapia , Plasticidade Celular , Células Cultivadas , Citocinas/metabolismo , Células Dendríticas/imunologia , Humanos , Tolerância Imunológica , Mediadores da Inflamação/metabolismo , Cadeias alfa de Integrinas/metabolismo , Lectinas Tipo C/metabolismo , Neoplasias Pulmonares/etiologia , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/terapia , Antígenos de Histocompatibilidade Menor/metabolismo , Receptores de Superfície Celular/metabolismo , Transdução de Sinais
17.
Front Immunol ; 9: 2489, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30455688

RESUMO

Toll-like receptor (TLR) agonists induce metabolic reprogramming, which is required for immune activation. We have investigated mechanisms that regulate metabolic adaptation upon TLR-stimulation in human blood DC subsets, CD1c+ myeloid DCs (mDCs) and plasmacytoid DCs (pDCs). We show that TLR-stimulation changes expression of genes regulating oxidative phosphorylation (OXPHOS) and glutamine metabolism in pDC. TLR-stimulation increases mitochondrial content and intracellular glutamine in an autophagy-dependent manner in pDC. TLR-induced glutaminolysis fuels OXPHOS in pDCs. Notably, inhibition of glutaminolysis and OXPHOS prevents pDC activation. Conversely, TLR-stimulation reduces mitochondrial content, OXPHOS activity and induces glycolysis in CD1c+ mDC. Inhibition of mitochondrial fragmentation or promotion of mitochondrial fusion impairs TLR-stimulation induced glycolysis and activation of CD1c+ mDCs. TLR-stimulation triggers BNIP3-dependent mitophagy, which regulates transcriptional activity of AMPKα1. BNIP3-dependent mitophagy is required for induction of glycolysis and activation of CD1c+ mDCs. Our findings reveal that TLR stimulation differentially regulates mitochondrial dynamics in distinct human DC subsets, which contributes to their activation.


Assuntos
Células Dendríticas/metabolismo , Mitocôndrias/metabolismo , Células Mieloides/metabolismo , Quinases Proteína-Quinases Ativadas por AMP , Antígenos CD1/metabolismo , Autofagia , Diferenciação Celular , Células Cultivadas , Reprogramação Celular , Citocinas/metabolismo , Células Dendríticas/imunologia , Glutamina/metabolismo , Glicólise , Glicoproteínas/metabolismo , Humanos , Imunidade , Proteínas de Membrana/metabolismo , Células Mieloides/imunologia , Fosforilação Oxidativa , Proteínas Quinases/genética , Proteínas Proto-Oncogênicas/metabolismo , Receptores Toll-Like/metabolismo
18.
Front Immunol ; 9: 744, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29755453

RESUMO

Dendritic cells (DCs) are major players for the induction of immune responses. Apart from plasmacytoid DCs (pDCs), human DCs can be categorized into two types of conventional DCs: CD141+ DCs (cDC1) and CD1c+ DCs (cDC2). Defining uniquely expressed surface markers on human immune cells is not only important for the identification of DC subpopulations but also a prerequisite for harnessing the DC subset-specific potential in immunomodulatory approaches, such as antibody-mediated antigen targeting. Although others identified CLEC9A as a specific endocytic receptor for CD141+ DCs, such a receptor for CD1c+ DCs has not been discovered, yet. By performing transcriptomic and flow cytometric analyses on human DC subpopulations from different lymphohematopoietic tissues, we identified CLEC10A (CD301, macrophage galactose-type C-type lectin) as a specific marker for human CD1c+ DCs. We further demonstrate that CLEC10A rapidly internalizes into human CD1c+ DCs upon binding of a monoclonal antibody directed against CLEC10A. The binding of a CLEC10A-specific bivalent ligand (the MUC-1 peptide glycosylated with N-acetylgalactosamine) is limited to CD1c+ DCs and enhances the cytokine secretion (namely TNFα, IL-8, and IL-10) induced by TLR 7/8 stimulation. Thus, CLEC10A represents not only a candidate to better define CD1c+ DCs-due to its high endocytic potential-CLEC10A also exhibits an interesting candidate receptor for future antigen-targeting approaches.


Assuntos
Antígenos CD1/imunologia , Células Dendríticas/imunologia , Glicoproteínas/imunologia , Lectinas Tipo C/imunologia , Adulto , Citocinas/imunologia , Humanos , Mucina-1/imunologia , Receptor 7 Toll-Like/imunologia , Receptor 8 Toll-Like/imunologia
19.
Hum Pathol ; 68: 147-153, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28414089

RESUMO

Myeloid malignancies showing CD141+ myeloid dendritic cell (MDC) differentiation have not been documented. Here, we describe a patient with juvenile myelomonocytic leukemia in which a prominent CD141+ cell population was identified most consistent with CD141+ MDCs based on phenotypic similarity with normal CD141+ MDCs. Molecular studies demonstrated a KRAS mutation. The findings from the spleen and bone marrow are described. This is the first well-documented demonstration of CD141+ MDC differentiation of a hematopoietic neoplasm.


Assuntos
Antígenos de Superfície/análise , Biomarcadores Tumorais/análise , Diferenciação Celular , Células Dendríticas/imunologia , Leucemia Mielomonocítica Juvenil/imunologia , Antígenos CD1/análise , Biomarcadores Tumorais/genética , Biópsia , Antígenos CD13/análise , Criança , Células Dendríticas/patologia , Citometria de Fluxo , Predisposição Genética para Doença , Glicoproteínas/análise , Humanos , Imuno-Histoquímica , Leucemia Mielomonocítica Juvenil/genética , Leucemia Mielomonocítica Juvenil/patologia , Leucemia Mielomonocítica Juvenil/terapia , Masculino , Mutação , Fenótipo , Proteínas Proto-Oncogênicas p21(ras)/genética , Trombomodulina
20.
J Leukoc Biol ; 102(4): 1055-1068, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28720687

RESUMO

Human 6-sulfo LacNac-positive (slan+) cells have been subject to a paradigm debate. They have previously been classified as a distinct dendritic cell (DC) subset. However, evidence has emerged that they may be more related to monocytes than to DCs. To gain deeper insight into the functional specialization of slan+ cells, we have compared them with both conventional myeloid DC subsets (CD1c+ and CD141+) in human peripheral blood (PB). With the use of genome-wide transcriptional profiling, as well as functional tests, we clearly show that slan+ cells form a distinct, non-DC-like population. They cluster away from both DC subsets, and their gene-expression profile evidently suggests involvement in distinct inflammatory processes. An extensive transcriptional meta-analysis confirmed the relationship of slan+ cells with the monocytic compartment rather than with DCs. From a functional perspective, their ability to prime CD4+ and CD8+ T cells is relatively low. Combined with the finding that "antigen presentation by MHC class II" is at the top of under-represented pathways in slan+ cells, this points to a minimal role in directing adaptive T cell immunity. Rather, the higher expression levels of complement receptors on their cell surface, together with their high secretion of IL-1ß and IL-6, imply a specific role in innate inflammatory processes, which is consistent with their recent identification as non-classical monocytes. This study extends our knowledge on DC/monocyte subset biology under steady-state conditions and contributes to our understanding of their role in immune-mediated diseases and their potential use in immunotherapeutic strategies.


Assuntos
Células Dendríticas/imunologia , Monócitos/imunologia , Transcrição Gênica/imunologia , Proteínas Supressoras de Tumor , Antígenos CD1/imunologia , Antígenos de Superfície/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/citologia , Perfilação da Expressão Gênica , Glicoproteínas/imunologia , Humanos , Interleucina-1beta/imunologia , Interleucina-6/imunologia , Monócitos/citologia , Trombomodulina
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa