Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
J Biomed Sci ; 31(1): 68, 2024 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-38992694

RESUMO

BACKGROUND: KRAS mutations frequently occur in cancers, particularly pancreatic ductal adenocarcinoma, colorectal cancer, and non-small cell lung cancer. Although KRASG12C inhibitors have recently been approved, effective precision therapies have not yet been established for all KRAS-mutant cancers. Many treatments for KRAS-mutant cancers, including epigenome-targeted drugs, are currently under investigation. Small ubiquitin-like modifier (SUMO) proteins are a family of small proteins covalently attached to and detached from other proteins in cells via the processes called SUMOylation and de-SUMOylation. We assessed whether SUMOylation inhibition was effective in KRAS-mutant cancer cells. METHODS: The efficacy of the first-in-class SUMO-activating enzyme E inhibitor TAK-981 (subasumstat) was assessed in multiple human and mouse KRAS-mutated cancer cell lines. A gene expression assay using a TaqMan array was used to identify biomarkers of TAK-981 efficacy. The biological roles of SUMOylation inhibition and subsequent regulatory mechanisms were investigated using immunoblot analysis, immunofluorescence assays, and mouse models. RESULTS: We discovered that TAK-981 downregulated the expression of the currently undruggable MYC and effectively suppressed the growth of MYC-expressing KRAS-mutant cancers across different tissue types. Moreover, TAK-981-resistant cells were sensitized to SUMOylation inhibition via MYC-overexpression. TAK-981 induced proteasomal degradation of MYC by altering the balance between SUMOylation and ubiquitination and promoting the binding of MYC and Fbxw7, a key factor in the ubiquitin-proteasome system. The efficacy of TAK-981 monotherapy in immunocompetent and immunodeficient mouse models using a mouse-derived CMT167 cell line was significant but modest. Since MAPK inhibition of the KRAS downstream pathway is crucial in KRAS-mutant cancer, we expected that co-inhibition of SUMOylation and MEK might be a good option. Surprisingly, combination treatment with TAK-981 and trametinib dramatically induced apoptosis in multiple cell lines and gene-engineered mouse-derived organoids. Moreover, combination therapy resulted in long-term tumor regression in mouse models using cell lines of different tissue types. Finally, we revealed that combination therapy complementally inhibited Rad51 and BRCA1 and accumulated DNA damage. CONCLUSIONS: We found that MYC downregulation occurred via SUMOylation inhibition in KRAS-mutant cancer cells. Our findings indicate that dual inhibition of SUMOylation and MEK may be a promising treatment for MYC-expressing KRAS-mutant cancers by enhancing DNA damage accumulation.


Assuntos
Dano ao DNA , Proteínas Proto-Oncogênicas p21(ras) , Sumoilação , Sumoilação/efeitos dos fármacos , Animais , Camundongos , Humanos , Linhagem Celular Tumoral , Dano ao DNA/efeitos dos fármacos , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Mutação , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética
2.
Immunity ; 42(6): 1048-61, 2015 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-26070482

RESUMO

Thymic antigen-presenting cells (APCs) such as dendritic cells and medullary thymic epithelial cells (mTECs) use distinct strategies of self-antigen expression and presentation to mediate central tolerance. The thymus also harbors B cells; whether they also display unique tolerogenic features and how they genealogically relate to peripheral B cells is unclear. Here, we found that Aire is expressed in thymic but not peripheral B cells. Aire expression in thymic B cells coincided with major histocompatibility class II (MHCII) and CD80 upregulation and immunoglobulin class-switching. These features were recapitulated upon immigration of naive peripheral B cells into the thymus, whereby this intrathymic licensing required CD40 signaling in the context of cognate interactions with autoreactive CD4(+) thymocytes. Moreover, a licensing-dependent neo-antigen selectively upregulated in immigrating B cells mediated negative selection through direct presentation. Thus, autoreactivity within the nascent T cell repertoire fuels a feed forward loop that endows thymic B cells with tolerogenic features.


Assuntos
Linfócitos B/fisiologia , Linfócitos T CD4-Positivos/imunologia , Antígenos CD40/metabolismo , Timo/imunologia , Fatores de Transcrição/metabolismo , Animais , Apresentação de Antígeno/genética , Autoantígenos/imunologia , Antígeno B7-1/genética , Antígeno B7-1/metabolismo , Antígenos CD40/genética , Diferenciação Celular/genética , Células Cultivadas , Tolerância Central/genética , Seleção Clonal Mediada por Antígeno/genética , Antígenos de Histocompatibilidade Classe II/genética , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Switching de Imunoglobulina/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Transdução de Sinais , Fatores de Transcrição/genética , Proteína AIRE
3.
Nano Lett ; 23(2): 619-628, 2023 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-36641798

RESUMO

Anti-spike neutralizing antibodies (S NAbs) have been developed for prevention and treatment against COVID-19. The nanoscopic characterization of the dynamic interaction between spike proteins and S NAbs remains difficult. By using high-speed atomic force microscopy (HS-AFM), we elucidate the molecular property of an S NAb and its interaction with spike proteins. The S NAb appeared as monomers with a Y conformation at low density and formed hexameric oligomers at high density. The dynamic S NAb-spike protein interaction at RBD induces neither RBD opening nor S1 subunit shedding. Furthermore, the interaction was stable at endosomal pH. These findings indicated that the S NAb could have a negligible risk of antibody-dependent enhancement. Dynamic movement of spike proteins on small extracellular vesicles (S sEV) resembled that on SARS-CoV-2. The sensitivity of variant S sEVs to S NAb could be evaluated using HS-AFM. Altogether, we demonstrate a nanoscopic assessment platform for evaluating the binding property of S NAbs.


Assuntos
COVID-19 , Humanos , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , Anticorpos Antivirais , Anticorpos Neutralizantes
4.
Anal Chem ; 95(38): 14159-14164, 2023 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-37709279

RESUMO

Extracellular vesicles (EVs), including exosomes, have been recognized as key mediators of intercellular communications through donor EV and recipient cell interaction. Until now, most studies have focused on the development of analytical tools to separate EVs and their applications for the molecular profiling of EV cargo. However, we lack a complete picture of the mechanism of EV uptake by the recipient cells. Here, we developed the TurboID-EV system with the engineered biotin ligase TurboID, tethered to the EV membrane, which allowed us to track the footprints of EVs during and after EV uptake by the proximity-dependent biotinylation of recipient cellular proteins. To analyze biotinylated recipient proteins from low amounts of input cells (corresponding to ∼10 µg of proteins), we developed an integrated proteomic workflow that combined stable isotope labeling with amino acids in cultured cells (SILAC), fluorescence-activated cell sorting, spintip-based streptavidin affinity purification, and mass spectrometry. Using this method, we successfully identified 456 biotinylated recipient proteins, including not only well-known proteins involved in endocytosis and macropinocytosis but also other membrane-associated proteins such as desmoplakin and junction plakoglobin. The TurboID-EV system should be readily applicable to various EV subtypes and recipient cell types, providing a promising tool to dissect the specificity of EV uptake mechanisms on a proteome-wide scale.


Assuntos
Exossomos , Vesículas Extracelulares , Proteômica , Endocitose , Proteínas de Membrana
5.
Pharm Res ; 40(4): 927-935, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36163411

RESUMO

PURPOSE: To inhibit the transmission of SARS-CoV-2, we developed engineered exosomes that were conjugated with anti-spike nanobodies and type I interferon ß (IFN-ß). We evaluated the efficacy and potency of nanobody-IFN-ß conjugated exosomes to treatment of SARS-CoV-2 infection. METHODS: Milk fat globule epidermal growth factor 8 (MFG-E8) is a glycoprotein that binds to phosphatidylserine (PS) exposed on the exosomes. We generated nanobody-IFN-ß conjugated exosomes by fusing an anti-spike nanobody and IFN-ß with MFG-E8. We used the SARS-CoV-2 pseudovirus with the spike of the D614G mutant that encodes ZsGreen to mimic the infection process of the SARS-CoV-2. The SARS-CoV-2 pseudovirus was infected with angiotensin-converting enzyme-2 (ACE2) expressing adenocarcinomic human alveolar basal epithelial cells (A549) or ACE2 expressing HEK-blue IFNα/ß cells in the presence of nanobody-IFN-ß conjugated exosomes. By assessing the expression of ZsGreen in target cells and the upregulation of interferon-stimulated genes (ISGs) in infected cells, we evaluated the anti-viral effects of nanobody-IFN-ß conjugated exosomes. RESULTS: We confirmed the anti-spike nanobody and IFN-ß expressions on the exosomes. Exosomes conjugated with nanobody-hIFN-ß inhibited the interaction between the spike protein and ACE2, thereby inhibiting the infection of host cells with SARS-CoV-2 pseudovirus. At the same time, IFN-ß was selectively delivered to SARS-CoV-2 infected cells, resulting in the upregulation of ISGs expression. CONCLUSION: Exosomes conjugated with nanobody-IFN-ß may provide potential benefits in the treatment of COVID-19 because of the cooperative anti-viral effects of the anti-spike nanobody and the IFN-ß.


Assuntos
COVID-19 , Exossomos , Humanos , SARS-CoV-2 , Enzima de Conversão de Angiotensina 2 , Interferon beta , Ligação Proteica , Anticorpos , Antivirais
6.
Eur J Immunol ; 49(2): 351-352, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30488950

RESUMO

We show that latently gammaherpesvirus-infected B cells are present in the thymus. This could result in a functional T-cell tolerance against certain viral epitopes. It is conceivable that also antigens from other viruses or pathogens may be conveyed to the thymus for their immune evasion.


Assuntos
Linfócitos B/imunologia , Infecções por Herpesviridae/imunologia , Evasão da Resposta Imune , Tolerância Imunológica , Rhadinovirus/imunologia , Timo/imunologia , Animais , Linfócitos B/patologia , Infecções por Herpesviridae/patologia , Camundongos , Timo/patologia
7.
J Immunol ; 201(10): 3051-3057, 2018 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-30333125

RESUMO

During inflammation, phagocytes release digestive enzymes from lysosomes to degrade harmful cells such as pathogens and tumor cells. However, the molecular mechanisms regulating this process are poorly understood. In this study, we identified myoferlin as a critical regulator of lysosomal exocytosis by mouse phagocytes. Myoferlin is a type II transmembrane protein with seven C2 domains in the cytoplasmic region. It localizes to lysosomes and mediates their fusion with the plasma membrane upon calcium stimulation. Myoferlin promotes the release of lysosomal contents, including hydrolytic enzymes, which increase cytotoxicity. These data demonstrate myoferlin's critical role in lysosomal exocytosis by phagocytes, providing novel insights into the mechanisms of inflammation-related cellular injuries.


Assuntos
Citotoxicidade Imunológica/imunologia , Lisossomos/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Musculares/metabolismo , Fagócitos/metabolismo , Animais , Exocitose/imunologia , Lisossomos/imunologia , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Musculares/imunologia , Células NIH 3T3 , Fagócitos/imunologia
8.
J Immunol ; 199(12): 3959-3971, 2017 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-29101311

RESUMO

Aire controls the fate of autoreactive thymocytes (i.e., clonal deletion or development into regulatory T cells [Tregs]) through transcriptional control of the expression of tissue-restricted self-antigens (TRAs) from medullary thymic epithelial cells (mTECs) and bone marrow (BM)-derived cells. Although TRAs expressed by mTECs and BM-derived cells are suggested to complement each other to generate a full spectrum of TRAs, little is known about the relative contribution of TRAs from each component for establishment of self-tolerance. Furthermore, the precise role of Aire in specific types of Aire-expressing APCs remains elusive. We have approached these issues by generating two different types of transgenic mouse (Tg) model, which express a prefixed model self-antigen driven by the insulin promoter or the Aire promoter. In the insulin-promoter Tg model, mTECs alone were insufficient for clonal deletion, and BM-derived APCs were required for this action by utilizing Ag transferred from mTECs. In contrast, mTECs alone were able to induce Tregs, although at a much lower efficiency in the absence of BM-derived APCs. Importantly, lack of Aire in mTECs, but not in BM-derived APCs, impaired both clonal deletion and production of Tregs. In the Aire-promoter Tg model, both mTECs and BM-derived APCs could independently induce clonal deletion without Aire, and production of Tregs was impaired by the lack of Aire in mTECs, but not in BM-derived APCs. These results suggest that the fate of autoreactive thymocytes together with the requirement for Aire depend on the cell types that express self-antigens and the types of APCs involved in tolerance induction.


Assuntos
Apresentação de Antígeno , Células Apresentadoras de Antígenos/imunologia , Autoantígenos/imunologia , Deleção Clonal/imunologia , Linfócitos T Reguladores/imunologia , Timo/imunologia , Fatores de Transcrição/imunologia , Animais , Autoantígenos/biossíntese , Autoantígenos/genética , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Feminino , Regulação da Expressão Gênica/imunologia , Técnicas de Introdução de Genes , Genes Sintéticos , Insulina/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Ovalbumina/biossíntese , Ovalbumina/genética , Ovalbumina/imunologia , Regiões Promotoras Genéticas , Ratos , Organismos Livres de Patógenos Específicos , Timo/citologia , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Transgenes , Proteína AIRE
9.
iScience ; 27(5): 109704, 2024 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-38680663

RESUMO

In addition to cross-presentation, cross-dressing plays an important role in the induction of CD8+ T cell immunity. In the process of cross-dressing, conventional dendritic cells (DCs) acquire major histocompatibility complex class I (MHCI) from other cells and subsequently prime CD8+ T cells via the pre-formed antigen-MHCI complexes without antigen processing. However, the mechanisms underlying the cross-dressing pathway, as well as the relative contributions of cross-presentation and cross-dressing to CD8+ T cell priming are not fully understood. Here, we demonstrate that DCs rapidly acquire MHCI-containing membrane fragments from dead cells via the phosphatidylserine recognition-dependent mechanism for cross-dressing. The MHCI dressing is enhanced by a TLR3 ligand polyinosinic-polycytidylic acid (polyI:C). Further, polyI:C promotes not only cross-presentation but also cross-dressing in vivo. Taken together, these results suggest that cross-dressing as well as cross-presentation is involved in inflammatory diseases associated with cell death and type I IFN production.

10.
Blood ; 117(9): 2640-8, 2011 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-21220748

RESUMO

Dendritic cells (DCs) are known to regulate immune responses by inducing both central and peripheral tolerance. DCs play a vital role in negative selection of developing thymocytes by deleting T cells with high-affinity for self-peptide-major histocompatibility complexes. In the periphery, DCs mediate peripheral tolerance by promoting regulatory T-cell development, induction of T-cell unresponsiveness, and deletion of activated T cells. We studied whether allogeneic DCs, obtained from bone marrow cultured with either Flt3L (FLDCs) or granulocyte-macrophage colony-stimulating factor (GMDCs), could induce allospecific central and peripheral tolerance after IV injection; B cells were used as a control. The results showed that only FLDCs reached the thymus after injection and that these cells induced both central and peripheral tolerance to donor major histocompatibility complexes. For central tolerance, injection of FLDCs induced antigen-specific clonal deletion of both CD8 and CD4 single-positive thymocytes. For peripheral tolerance, injection of FLDCs induced donor-specific T-cell unresponsiveness and prolonged survival of donor-derived skin grafts. Tolerance induction by adoptive transfer of FLDCs could be a useful approach for promoting graft acceptance after organ transplantation.


Assuntos
Células Dendríticas/citologia , Células Dendríticas/imunologia , Sobrevivência de Enxerto/imunologia , Tolerância Imunológica/imunologia , Transplante de Pele/imunologia , Transferência Adotiva , Animais , Movimento Celular , Proliferação de Células , Células Clonais , Epitopos/imunologia , Injeções , Proteínas de Membrana/metabolismo , Camundongos , Células da Side Population/citologia , Células da Side Population/imunologia , Linfócitos T/citologia , Linfócitos T/imunologia , Timo/citologia , Fatores de Tempo , Transplante Homólogo
11.
J Mater Sci Mater Med ; 23(10): 2399-412, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22588504

RESUMO

The use of an "over 1000-nm near-infrared (NIR) in vivo fluorescence bioimaging" system based on lanthanide containing inorganic nanostructures emitting in the visible and NIR range under 980-nm excitation is proposed. It may overcome problems of currently used biomarkers including color fading, phototoxicity and scattering. Gd(2)O(3):Er(3+),Yb(3+) nanoparticles and nanorods showing upconversion and NIR emission are synthesized and their cytotoxic behavior is investigated by incubation with B-cell hybridomas and macrophages. Surface modification with PEG-b-PAAc provides the necessary chemical durability reducing the release of toxic Gd(3+) ions. NIR fluorescence microscopy is used to investigate the suitability of the nanostructures as NIR-NIR biomarkers. The in vitro uptake of bare and modified nanostructures by macrophages is investigated by confocal laser scanning microscopy. In vivo investigations revealed nanostructures in liver, lung, kidneys and spleen a few hours after injection into mice, while most of the nanostructures have been removed from the body after 24 h.


Assuntos
Érbio/química , Gadolínio/química , Nanoestruturas , Espectroscopia de Luz Próxima ao Infravermelho/métodos , Itérbio/química , Animais , Materiais Biocompatíveis , Linhagem Celular , Sobrevivência Celular , Érbio/farmacocinética , Gadolínio/farmacocinética , Técnicas In Vitro , Camundongos , Microscopia Eletrônica de Varredura , Difração de Pó , Propriedades de Superfície , Distribuição Tecidual , Itérbio/farmacocinética
12.
J Extracell Vesicles ; 11(11): e12275, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36317784

RESUMO

Small extracellular vesicles (sEVs) play a crucial role in local and distant cell communication. The intrinsic properties of sEVs make them compatible biomaterials for drug delivery, vaccines, and theranostic nanoparticles. Although sEV proteomics have been robustly studied, a direct instantaneous assessment of sEV structure dynamics remains difficult. Here, we use the high-speed atomic force microscopy (HS-AFM) to evaluate nanotopological changes of sEVs with respect to different physicochemical stresses including thermal stress, pH, and osmotic stress. The sEV structure is severely altered at high-temperature, high-pH, or hypertonic conditions. Surprisingly, the spherical shape of the sEVs is maintained in acidic or hypotonic environments. Real-time observation by HS-AFM imaging reveals an irreversible structural change in the sEVs during transition of pH or osmolarity. HS-AFM imaging provides both qualitative and quantitative data at high spatiotemporal resolution (nanoscopic and millisecond levels). In summary, our study demonstrates the feasibility of HS-AFM for structural characterization and assessment of nanoparticles.


Assuntos
Vesículas Extracelulares , Microscopia de Força Atômica/métodos
13.
Nat Commun ; 11(1): 4607, 2020 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-32929081

RESUMO

Drug tolerance is the basis for acquired resistance to epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) including osimertinib, through mechanisms that still remain unclear. Here, we show that while AXL-low expressing EGFR mutated lung cancer (EGFRmut-LC) cells are more sensitive to osimertinib than AXL-high expressing EGFRmut-LC cells, a small population emerge osimertinib tolerance. The tolerance is mediated by the increased expression and phosphorylation of insulin-like growth factor-1 receptor (IGF-1R), caused by the induction of its transcription factor FOXA1. IGF-1R maintains association with EGFR and adaptor proteins, including Gab1 and IRS1, in the presence of osimertinib and restores the survival signal. In AXL-low-expressing EGFRmut-LC cell-derived xenograft and patient-derived xenograft models, transient IGF-1R inhibition combined with continuous osimertinib treatment could eradicate tumors and prevent regrowth even after the cessation of osimertinib. These results indicate that optimal inhibition of tolerant signals combined with osimertinib may dramatically improve the outcome of EGFRmut-LC.


Assuntos
Acrilamidas/uso terapêutico , Compostos de Anilina/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Mutação/genética , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Receptor IGF Tipo 1/antagonistas & inibidores , Acrilamidas/farmacologia , Idoso de 80 Anos ou mais , Compostos de Anilina/farmacologia , Animais , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Receptores ErbB/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Fator 3-alfa Nuclear de Hepatócito/metabolismo , Humanos , Imidazóis/farmacologia , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Modelos Biológicos , Fosforilação/efeitos dos fármacos , Pirazinas/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Regulação para Cima/efeitos dos fármacos , Receptor Tirosina Quinase Axl
14.
Sci Rep ; 9(1): 4695, 2019 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-30886174

RESUMO

Gene correction of induced pluripotent stem cells (iPSCs) has therapeutic potential for treating homozygous familial hypercholesterolemia (HoFH) associated with low-density lipoprotein (LDL) receptor (LDLR) dysfunction. However, few data exist regarding the functional recovery and immunogenicity of LDLR gene-corrected iPSC-derived hepatocyte-like cells (HLCs) obtained from an HoFH patient. Therefore, we generated iPSC-derived HLCs from an HoFH patient harbouring a point mutation (NM_000527.4:c.901 G > T) in exon 6 of LDLR, and examined their function and immunogenicity. From the patient's iPSCs, one homozygous gene-corrected HoFH-iPSC clone and two heterozygous clones were generated using the CRISPR/Cas9 method. Both types of iPSC-derived HLCs showed recovery of the function of LDL uptake in immunofluorescence staining analysis. Furthermore, these gene-corrected iPSC-derived HLCs showed little immunogenicity against the patient's peripheral blood mononuclear cells in a cell-mediated cytotoxicity assay. These results demonstrate that LDL uptake of iPSC-derived HLCs from HoFH can be restored by gene correction without the appearance of further immunogenicity, suggesting that gene-corrected iPSC-derived HLCs are applicable to the treatment of HoFH.


Assuntos
Terapia Biológica/métodos , Terapia Genética/métodos , Hepatócitos/citologia , Hiperlipoproteinemia Tipo II/imunologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Lipoproteínas LDL/metabolismo , Diferenciação Celular , Linhagem Celular , Células Cultivadas , LDL-Colesterol/metabolismo , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Citotoxicidade Imunológica , Hepatócitos/metabolismo , Homozigoto , Humanos , Hiperlipoproteinemia Tipo II/genética , Células-Tronco Pluripotentes Induzidas/transplante , Lipoproteínas LDL/genética , Mutação/genética
15.
J Exp Med ; 216(5): 1027-1037, 2019 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-30918005

RESUMO

The autoimmune regulator (Aire) serves an essential function for T cell tolerance by promoting the "promiscuous" expression of tissue antigens in thymic epithelial cells. Aire is also detected in rare cells in peripheral lymphoid organs, but the identity of these cells is poorly understood. Here, we report that Aire protein-expressing cells in lymph nodes exhibit typical group 3 innate lymphoid cell (ILC3) characteristics such as lymphoid morphology, absence of "classical" hematopoietic lineage markers, and dependence on RORγt. Aire+ cells are more frequent among lineage-negative RORγt+ cells of peripheral lymph nodes as compared with mucosa-draining lymph nodes, display a unique Aire-dependent transcriptional signature, express high surface levels of MHCII and costimulatory molecules, and efficiently present an endogenously expressed model antigen to CD4+ T cells. These findings define a novel type of ILC3-like cells with potent APC features, suggesting that these cells serve a function in the control of T cell responses.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Linfonodos/citologia , Linfócitos/imunologia , Linfócitos/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Animais , Antígenos CD11/metabolismo , Molécula de Adesão da Célula Epitelial/metabolismo , Regulação da Expressão Gênica , Antígenos de Histocompatibilidade Classe II/metabolismo , Imunidade Inata , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Fenótipo , Transcrição Gênica , Proteína AIRE
16.
Front Immunol ; 6: 376, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26257742

RESUMO

Central T cell tolerance is believed to be mainly induced by thymic dendritic cells and medullary thymic epithelial cells. The thymus also harbors substantial numbers of B cells. These may arise though intrathymic B lymphopoiesis or immigration from the bloodstream. Importantly, and in contrast to resting "mainstream" B cells in the periphery, thymic B cells display elevated levels of MHC class II and constitutively express CD80. Arguably, their most unexpected feature is the expression of autoimmune regulator. These unique features of thymic B cells result from a licensing process that involves cross-talk with CD4 single-positive T cells and CD40 signaling. Together, these recent findings suggest that B cells play a more prominent role as thymic APCs than previously appreciated.

17.
Acta Biomater ; 9(1): 4734-43, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22963845

RESUMO

Bioimaging is an important diagnostic tool in the investigation and visualization of biological phenomena in cells and in medicine. In this context, up-converting Gd(2)O(3):Er(3+),Yb(3+) nanostructures (nanoparticles, nanorods) have been synthesized by precipitation methods and hydrothermal synthesis. Independent of size and morphology, Gd(2)O(3):Er(3+),Yb(3+) powders show up-conversion (550 nm, 670 nm) and near-infrared emission (1.5 µm) upon 980 nm excitation, which makes these structures interesting for application as biomarkers. With regard to their potential application in bioimaging, cytotoxicity is an important aspect and is strongly affected by the physico-chemical properties of the investigated nanostructures. Therefore, the cytotoxic effect of bare and poly(ethylene glycol)-b-poly(acrylic acid) block co-polymer-modified nanostructures on non-phagocytic and phagocytic cells (B-cell hybridoma cells and macrophages) was investigated. The observed cytotoxic behavior in the case of macrophages incubated with bare nanostructures was assigned to the poor chemical durability of gadolinium oxide, but could be overcome by surface modification.


Assuntos
Gadolínio/química , Nanoestruturas , Espectroscopia de Luz Próxima ao Infravermelho/métodos , Microscopia Eletrônica de Varredura , Difração de Pó
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA