RESUMO
BACKGROUND: Peanut is a potent inducer of proallergenic TH2 responses in susceptible individuals. Antigen-presenting cells (APCs) including dendritic cells and monocytes instruct naive T cells to differentiate into various effector cells, determining immune responses such as allergy and tolerance. OBJECTIVE: We sought to detect peanut protein (PN)-induced changes in gene expression in human myeloid dendritic cells (mDCs) and monocytes, identify signaling receptors that mediate these changes, and assess how PN-induced genes in mDCs impact their ability to promote T-cell differentiation. METHODS: mDCs, monocytes, and naive CD4+ T cells were isolated from blood bank donors and peanut-allergic patients. APCs were incubated with PN and other stimulants, and gene expression was measured using microarray and RT quantitative PCR. To assess T-cell differentiation, mDCs were cocultured with naive TH cells. RESULTS: PN induced a unique gene expression profile in mDCs, including the gene that encodes retinaldehyde dehydrogenase 2 (RALDH2), a rate-limiting enzyme in the retinoic acid (RA)-producing pathway. Stimulation of mDCs with PN also induced a 7-fold increase in the enzymatic activity of RALDH2. Blocking antibodies against Toll-like receptor (TLR)1/TLR2, as well as small interfering RNA targeting TLR1/TLR2, reduced the expression of RALDH2 in PN-stimulated APCs by 70%. Naive TH cells cocultured with PN-stimulated mDCs showed an RA-dependent 4-fold increase in production of IL-5 and expression of integrin α4ß7. CONCLUSIONS: PN induces RALDH2 in human APCs by signaling through the TLR1/TLR2 heterodimer. This leads to production of RA, which acts on TH cells to induce IL-5 and gut-homing integrin. RALDH2 induction by PN in APCs and RA-promoted TH2 differentiation could be an important factor determining allergic responses to peanut.
Assuntos
Família Aldeído Desidrogenase 1/imunologia , Células Apresentadoras de Antígenos/imunologia , Arachis/imunologia , Retinal Desidrogenase/imunologia , Células Th2/imunologia , Linfócitos T CD4-Positivos/imunologia , Diferenciação Celular/imunologia , Células Cultivadas , Células Dendríticas/imunologia , Células HEK293 , Humanos , Hipersensibilidade/imunologia , Ativação Linfocitária/imunologia , Monócitos/imunologia , Tretinoína/imunologiaRESUMO
Pancreatic neuroendocrine tumors (PanNETs) are a type of pancreatic cancer with limited therapeutic options. Consequently, most patients with advanced disease die from tumor progression. Current evidence indicates that a subset of cancer cells is responsible for tumor development, metastasis, and recurrence, and targeting these tumor-initiating cells is necessary to eradicate tumors. However, tumor-initiating cells and the biological processes that promote pathogenesis remain largely uncharacterized in PanNETs. Here we profile primary and metastatic tumors from an index patient and demonstrate that MET proto-oncogene activation is important for tumor growth in PanNET xenograft models. We identify a highly tumorigenic cell population within several independent surgically acquired PanNETs characterized by increased cell-surface protein CD90 expression and aldehyde dehydrogenase A1 (ALDHA1) activity, and provide in vitro and in vivo evidence for their stem-like properties. We performed proteomic profiling of 332 antigens in two cell lines and four primary tumors, and showed that CD47, a cell-surface protein that acts as a "don't eat me" signal co-opted by cancers to evade innate immune surveillance, is ubiquitously expressed. Moreover, CD47 coexpresses with MET and is enriched in CD90(hi)cells. Furthermore, blocking CD47 signaling promotes engulfment of tumor cells by macrophages in vitro and inhibits xenograft tumor growth, prevents metastases, and prolongs survival in vivo.
Assuntos
Tumores Neuroendócrinos , Neoplasias Pancreáticas , Evasão Tumoral , Família Aldeído Desidrogenase 1 , Animais , Antígeno CD47/imunologia , Feminino , Humanos , Isoenzimas/imunologia , Masculino , Camundongos Endogâmicos NOD , Camundongos SCID , Metástase Neoplásica , Proteínas de Neoplasias/imunologia , Tumores Neuroendócrinos/imunologia , Tumores Neuroendócrinos/patologia , Tumores Neuroendócrinos/terapia , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/terapia , Proto-Oncogene Mas , Retinal Desidrogenase/imunologia , Antígenos Thy-1/imunologia , Ensaios Antitumorais Modelo de XenoenxertoRESUMO
The vitamin A metabolite retinoic acid (RA) has been reported to suppress Th1 responses and enhance Th2 responses. Here, we investigated whether differences in vitamin A metabolism could underlie the differences between C57BL/6 and BALB/c mice, which are reportedly seen as Th1 and Th2 responders, respectively. BALB/c mice were shown to have higher intestinal epithelial expression of RALDH1 (where RALDH is retinaldehyde dehydrogenase), and, consequently, higher RALDH activity in MLN-DCs, leading to an increased ability to induce IgA class switching in B cells. Furthermore, within BALB/c mice, induction of IgA secretion as well as increased accumulation of regulatory T cells (Treg) in the intestinal lamina propria was observed. Additionally, as BALB/c mice are more resistant to dextran sulphate sodium (DSS) induced colitis, mice that lacked vitamin A in their diet had a more severe form of DSS-induced colitis compared to control mice. Therefore, the level of RA production and consequently the degree of RA-mediated signaling is crucial for the efficiency of the mucosal immune system.
Assuntos
Colite/imunologia , Imunidade nas Mucosas , Intestinos/imunologia , Isoenzimas/imunologia , Mucosa/imunologia , Retinal Desidrogenase/imunologia , Vitamina A/metabolismo , Família Aldeído Desidrogenase 1 , Animais , Colite/induzido quimicamente , Colite/metabolismo , Colite/patologia , Sulfato de Dextrana , Expressão Gênica , Imunoglobulina A/genética , Imunoglobulina A/imunologia , Imunoglobulina A/metabolismo , Switching de Imunoglobulina , Mucosa Intestinal/metabolismo , Intestinos/patologia , Isoenzimas/genética , Isoenzimas/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Mucosa/metabolismo , Mucosa/patologia , Retinal Desidrogenase/genética , Retinal Desidrogenase/metabolismo , Transdução de Sinais , Especificidade da Espécie , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/patologia , Células Th1/imunologia , Células Th1/metabolismo , Células Th1/patologia , Células Th2/imunologia , Células Th2/metabolismo , Células Th2/patologia , Vitamina A/administração & dosagemRESUMO
Although the vitamin A metabolite retinoic acid (RA) plays a critical role in immune function, RA synthesis during infection is poorly understood. Here, we show that retinal dehydrogenases (Raldh), required for the synthesis of RA, are induced during a retinoid-dependent type-2 immune response elicited by Schistosoma mansoni infection, but not during a retinoid-independent anti-viral immune response. Vitamin A deficient mice have a selective defect in T(H)2 responses to S. mansoni, but retained normal LCMV specific T(H)1 responses. A combination of in situ imaging, intra-vital imaging, and sort purification revealed that alternatively activated macrophages (AAMφ) express high levels of Raldh2 during S. mansoni infection. IL-4 induces Raldh2 expression in bone marrow-derived macrophages in vitro and peritoneal macrophages in vivo. Finally, in vivo derived AAMφ have an enhanced capacity to induce Foxp3 expression in CD4+ cells through an RA dependent mechanism, especially in combination with TGF-ß. The regulation of Raldh enzymes during infection is pathogen specific and reflects differential requirements for RA during effector responses. Specifically, AAMφ are an inducible source of RA synthesis during helminth infections and T(H)2 responses that may be important in regulating immune responses.
Assuntos
Regulação Enzimológica da Expressão Gênica/imunologia , Ativação de Macrófagos/imunologia , Macrófagos Peritoneais/imunologia , Retinal Desidrogenase/imunologia , Schistosoma mansoni/imunologia , Esquistossomose mansoni/imunologia , Regulação para Cima/imunologia , Animais , Células Cultivadas , Fatores de Transcrição Forkhead/biossíntese , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/imunologia , Regulação Enzimológica da Expressão Gênica/genética , Ativação de Macrófagos/genética , Camundongos , Camundongos Knockout , Retinal Desidrogenase/biossíntese , Retinal Desidrogenase/genética , Schistosoma mansoni/metabolismo , Esquistossomose mansoni/enzimologia , Esquistossomose mansoni/genética , Células Th1/imunologia , Células Th2/imunologia , Regulação para Cima/genéticaRESUMO
Purpose: Rapidly progressing cataract is one of the ocular manifestations in leptospiral uveitis patients. We examined whether molecular mimicry between the leptospira antigens and lens proteins exists that could result in cataract in these patients.Methods: Immunoblot analysis using patient sera was done with proteins from normal lens and cataract lens from leptospiral uveitis patients and the cross-reacting lens proteins were identified by mass spectrometry analysis.Results: Retinal dehydrogenase 1 and crystallins (α-B, α-A2, ß-B2), were recognized by the antibodies in the serum of leptospiral uveitis patients. And, retinal dehydrogenase 1 is homologous to the leptospiral protein, betaine aldehyde dehydrogenase.Conclusions: Leptospiral uveitis patient serum contains antibodies that cross-react with multiple lens proteins that have a role in maintaining lens transparency. And, these antibodies could act as a potential trigger for cataractogenesis.
Assuntos
Betaína-Aldeído Desidrogenase/imunologia , Catarata/imunologia , Cristalino/enzimologia , Leptospira/enzimologia , Leptospirose/imunologia , Mimetismo Molecular/fisiologia , Retinal Desidrogenase/imunologia , Uveíte/imunologia , Sequência de Aminoácidos , Anticorpos Antibacterianos/sangue , Antígenos de Bactérias/imunologia , Catarata/microbiologia , Reações Cruzadas/imunologia , Eletroforese em Gel de Poliacrilamida , Ensaio de Imunoadsorção Enzimática , Infecções Oculares Bacterianas/imunologia , Infecções Oculares Bacterianas/microbiologia , Humanos , Immunoblotting , Leptospirose/microbiologia , Espectrometria de Massas , Dados de Sequência Molecular , Uveíte/microbiologiaRESUMO
Glioblastoma (GBM) is the most aggressive malignant glioma. Therapeutic targeting of GBM is made more difficult due to its heterogeneity, resistance to treatment, and diffuse infiltration into the brain parenchyma. Better understanding of the tumor microenvironment should aid in finding more effective management of GBM. GBM-associated macrophages (GAM) comprise up to 30% of the GBM microenvironment. Therefore, exploration of GAM activity/function and their specific markers are important for developing new therapeutic agents. In this study, we identified and evaluated the expression of ALDH1A2 in the GBM microenvironment, and especially in M2 GAM, though it is also expressed in reactive astrocytes and multinucleated tumor cells. We demonstrated that M2 GAM highly express ALDH1A2 when compared to other ALDH1 family proteins. Additionally, GBM samples showed higher expression of ALDH1A2 when compared to low-grade gliomas (LGG), and this expression was increased upon tumor recurrence both at the gene and protein levels. We demonstrated that the enzymatic product of ALDH1A2, retinoic acid (RA), modulated the expression and activity of MMP-2 and MMP-9 in macrophages, but not in GBM tumor cells. Thus, the expression of ALDH1A2 may promote the progressive phenotype of GBM.
Assuntos
Família Aldeído Desidrogenase 1/metabolismo , Regulação Neoplásica da Expressão Gênica , Glioblastoma/patologia , Macrófagos/imunologia , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Retinal Desidrogenase/metabolismo , Família Aldeído Desidrogenase 1/genética , Família Aldeído Desidrogenase 1/imunologia , Apoptose , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Movimento Celular , Proliferação de Células , Glioblastoma/genética , Glioblastoma/imunologia , Glioblastoma/metabolismo , Humanos , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 9 da Matriz/genética , Retinal Desidrogenase/genética , Retinal Desidrogenase/imunologia , Tretinoína/metabolismo , Células Tumorais Cultivadas , Microambiente TumoralRESUMO
Dendritic cells (DCs) promote T-cell mediated tolerance to self-antigens and induce inflammation to innocuous-antigens. This dual potential makes DCs fundamental players in inflammatory disorders. Evidence from inflammatory colitis mouse models and inflammatory bowel diseases (IBD) patients indicated that gut inflammation in IBD is driven mainly by T-helper-1 (Th1) and Th17 cells, suggesting an essential role for DCs in the development of IBD. Here we show that GSK-J4, a selective inhibitor of the histone demethylase JMJD3/UTX, attenuated inflammatory colitis by reducing the inflammatory potential and increasing the tolerogenic features of DCs. Mechanistic analyses revealed that GSK-J4 increased activating epigenetic signals while reducing repressive marks in the promoter of retinaldehyde dehydrogenase isoforms 1 and 3 in DCs, enhancing the production of retinoic acid. This, in turn, has an impact on regulatory T cells (Treg) increasing their lineage stability and gut tropism as well as potentiating their suppressive activity. Our results open new avenues for the treatment of IBD patients.
Assuntos
Benzazepinas/farmacologia , Colite/imunologia , Células Dendríticas/imunologia , Doenças Inflamatórias Intestinais/imunologia , Pirimidinas/farmacologia , Tretinoína/imunologia , Família Aldeído Desidrogenase 1/genética , Família Aldeído Desidrogenase 1/imunologia , Animais , Colite/tratamento farmacológico , Colite/genética , Colite/patologia , Células Dendríticas/patologia , Doenças Inflamatórias Intestinais/tratamento farmacológico , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/patologia , Camundongos , Camundongos Knockout , Retinal Desidrogenase/genética , Retinal Desidrogenase/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/patologia , Células Th1/imunologia , Células Th1/patologia , Células Th17/imunologia , Células Th17/patologiaRESUMO
RDH12 codes for a member of the family of short-chain alcohol dehydrogenases/reductases proposed to function in the visual cycle that supplies the chromophore 11-cis retinal to photoreceptor cells. Mutations in RDH12 cause severe and progressive childhood onset autosomal-recessive retinal dystrophy, including Leber congenital amaurosis. We generated Rdh12 knockout mice, which exhibited grossly normal retinal histology at 10 months of age. Levels of all-trans and 11-cis retinoids in dark- and light-adapted animals and scotopic and photopic electroretinogram (ERG) responses were similar to those for the wild type, as was recovery of the ERG response following bleaching, for animals matched for an Rpe65 polymorphism (p.L450M). Lipid peroxidation products and other measures of oxidative stress did not appear to be elevated in Rdh12(-/-) animals. RDH12 was localized to photoreceptor inner segments and the outer nuclear layer in both mouse and human retinas by immunohistochemistry. The present findings, together with those of earlier studies showing only minor functional deficits in mice deficient for Rdh5, Rdh8, or Rdh11, suggest that the activity of any one isoform is not rate limiting in the visual response.
Assuntos
Marcação de Genes , Retinal Desidrogenase/genética , Retinal Desidrogenase/metabolismo , Visão Ocular/fisiologia , Oxirredutases do Álcool , Animais , Eletrorretinografia , Regulação da Expressão Gênica , Humanos , Imuno-Histoquímica , Camundongos , Estresse Oxidativo , Células Fotorreceptoras de Vertebrados/citologia , Células Fotorreceptoras de Vertebrados/enzimologia , Células Fotorreceptoras de Vertebrados/ultraestrutura , Transporte Proteico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Retinal Desidrogenase/deficiência , Retinal Desidrogenase/imunologia , Retinoides/análise , Visão Ocular/genéticaRESUMO
Retinal dehydrogenase (RALDH) enzymatic activities catalyze the conversion of vitamin A to its metabolite Retinoic acid (RA) in intestinal dendritic cells (DCs) and promote immunological tolerance. However, precise understanding of the exogenous factors that act as initial trigger of RALDH activity in these cells is still evolving. By using germ-free (GF) mice raised on an antigen free (AF) elemental diet, we find that certain components in diet are critically required to establish optimal RALDH expression and activity, most prominently in small intestinal CD103+CD11b+ DCs (siLP-DCs) right from the beginning of their lives. Surprisingly, systematic screens using modified diets devoid of individual dietary components indicate that proteins, starch and minerals are dispensable for this activity. On the other hand, in depth comparison between subtle differences in dietary composition among different dietary regimes reveal that adequate glucose concentration in diet is a critical determinant for establishing RALDH activity specifically in siLP-DCs. Consequently, pre-treatment of siLP-DCs, and not mesenteric lymph node derived MLNDCs with glucose, results in significant enhancement in the in vitro generation of induced Regulatory T (iTreg) cells. Our findings reveal previously underappreciated role of dietary glucose concentration in establishing regulatory properties in intestinal DCs, thereby extending a potential therapeutic module against intestinal inflammation.
Assuntos
Antígenos CD/metabolismo , Antígeno CD11b/metabolismo , Células Dendríticas/efeitos dos fármacos , Açúcares da Dieta/administração & dosagem , Glucose/administração & dosagem , Cadeias alfa de Integrinas/metabolismo , Intestino Delgado/efeitos dos fármacos , Retinal Desidrogenase/metabolismo , Ração Animal , Animais , Antígenos CD/imunologia , Antígeno CD11b/imunologia , Células Cultivadas , Técnicas de Cocultura , Células Dendríticas/enzimologia , Células Dendríticas/imunologia , Cadeias alfa de Integrinas/imunologia , Intestino Delgado/enzimologia , Intestino Delgado/imunologia , Camundongos Endogâmicos C57BL , Retinal Desidrogenase/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismoRESUMO
Epidemiological evidence correlates low serum vitamin A (retinol) levels with increased susceptibility to active tuberculosis (TB); however, retinol is biologically inactive and must be converted into its bioactive form, all-trans retinoic acid (ATRA). Given that ATRA triggers a Niemann-Pick type C2 (NPC2)-dependent antimicrobial response against Mycobacterium tuberculosis, we investigated the mechanism by which the immune system converts retinol into ATRA at the site of infection. We demonstrate that granulocyte-macrophage colony-stimulating factor (GM-CSF)-derived dendritic cells (DCs), but not macrophages, express enzymes in the vitamin A metabolic pathway, including aldehyde dehydrogenase 1 family, member a2 (ALDH1A2) and short-chain dehydrogenase/reductase family, member 9 (DHRS9), enzymes capable of the two-step conversion of retinol into ATRA, which is subsequently released from the cell. Additionally, mRNA and protein expression levels of ALDH1A2 and DC marker CD1B were lower in tuberculosis lung tissues than in normal lung. The conditioned medium from DCs cultured with retinol stimulated antimicrobial activity from M. tuberculosis-infected macrophages, as well as the expression of NPC2 in monocytes, which was blocked by specific inhibitors, including retinoic acid receptor inhibitor (RARi) or N,N-diethylaminobenzaldehyde (DEAB), an ALDH1A2 inhibitor. These results indicate that metabolism of vitamin A by DCs transactivates macrophage antimicrobial responses.IMPORTANCE Tuberculosis (TB) is the leading cause of death by a single infectious agent worldwide. One factor that contributes to the success of the microbe is the deficiency in immunomodulatory nutrients, such as vitamin A (retinol), which are prevalent in areas where TB is endemic. Clinical trials show that restoration of systemic retinol levels in active TB patients is ineffective in mitigating the disease; however, laboratory studies demonstrate that activation of the vitamin A pathway in Mycobacterium tuberculosis-infected macrophages triggers an antimicrobial response. Therefore, the goal of this study was to determine the link between host retinol levels and retinoic acid-mediated antimicrobial responses against M. tuberculosis By combining established in vitro models with in situ studies of lung tissue from TB patients, this study demonstrates that the innate immune system utilizes transcellular metabolism leading to activation between dendritic cells and macrophages as a means to combat the pathogen.
Assuntos
Células Dendríticas/enzimologia , Células Dendríticas/imunologia , Mycobacterium tuberculosis/imunologia , Vitamina A/metabolismo , 3-Hidroxiesteroide Desidrogenases/genética , 3-Hidroxiesteroide Desidrogenases/imunologia , Adulto , Família Aldeído Desidrogenase 1/genética , Família Aldeído Desidrogenase 1/imunologia , Células Cultivadas , Meios de Cultivo Condicionados/química , Células Dendríticas/microbiologia , Humanos , Pulmão/microbiologia , Macrófagos/enzimologia , Macrófagos/imunologia , Macrófagos/microbiologia , Retinal Desidrogenase/genética , Retinal Desidrogenase/imunologia , Tuberculose/microbiologiaRESUMO
Mucosal surfaces are the primary point of entry for many infectious agents and mucosal immune responses serve as the primary defense to these pathogens. In order to mount an effective mucosal immune response, it is important to induce T cell homing to mucosal surfaces. Conventional vaccine adjuvants induce strong systemic immunity but often fail to produce mucosal immunity. We have developed an oil-in-water nanoemulsion (NE) adjuvant that provides mucosal immunity and efficient protection against mucosal pathogens when administered as part of an intranasal vaccine. In the present study, we demonstrate that intranasal immunization with NE indirectly activates the retinaldehyde dehydrogenase (RALDH) activity in dendritic cells through epithelial cell activity leading to SIgA as well as potent cellular responses and expression of α4ß7 and CCR9 gut homing receptors on T cells. Confirming these findings, ex-vivo stimulation of splenocytes from NE nasally immunized animals showed increase in Th1/Th17 cytokines while suppressing Th2 responses. In examining mechanisms underlying this activation NE activated RALDH via MyD88 dependent pathways in DCs but did not activate the retinoic acid receptor directly. These results suggest that RALDH immune activities can be achieved by epithelial activation without direct RAR activation, which has significant implications for understanding mucosal immunity and the design of mucosal vaccines.
Assuntos
Adjuvantes Imunológicos/farmacologia , Células Dendríticas/imunologia , Imunidade nas Mucosas/efeitos dos fármacos , Fator 88 de Diferenciação Mieloide/imunologia , Nanoestruturas , Retinal Desidrogenase/imunologia , Transdução de Sinais/efeitos dos fármacos , Administração Intranasal , Animais , Linhagem Celular , Emulsões , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/imunologia , Camundongos , Transdução de Sinais/imunologiaRESUMO
In order for vaccines to induce efficacious immune responses against mucosally transmitted pathogens, such as HIV-1, activated lymphocytes must efficiently migrate to and enter targeted mucosal sites. We have previously shown that all-trans retinoic acid (ATRA) can be used as a vaccine adjuvant to enhance mucosal CD8+ T cell responses during vaccination and improve protection against mucosal viral challenge. However, the ATRA formulation is incompatible with most recombinant vaccines, and the teratogenic potential of ATRA at high doses limits its usage in many clinical settings. We hypothesized that increasing in vivo production of retinoic acid (RA) during vaccination with a DNA vector expressing retinaldehyde dehydrogenase 2 (RALDH2), the rate-limiting enzyme in RA biosynthesis, could similarly provide enhanced programming of mucosal homing to T cell responses while avoiding teratogenic effects. Administration of a RALDH2- expressing plasmid during immunization with a HIVgag DNA vaccine resulted in increased systemic and mucosal CD8+ T cell numbers with an increase in both effector and central memory T cells. Moreover, mice that received RALDH2 plasmid during DNA vaccination were more resistant to intravaginal challenge with a recombinant vaccinia virus expressing the same HIVgag antigen (VACVgag). Thus, RALDH2 can be used as an alternative adjuvant to ATRA during DNA vaccination leading to an increase in both systemic and mucosal T cell immunity and better protection from viral infection at mucosal sites.
Assuntos
Vacinas contra a AIDS/imunologia , Adjuvantes Imunológicos , Imunidade nas Mucosas , Retinal Desidrogenase/imunologia , Vacinas de DNA/imunologia , Vacinas contra a AIDS/administração & dosagem , Vacinas contra a AIDS/genética , Família Aldeído Desidrogenase 1 , Animais , Linfócitos T CD8-Positivos/imunologia , Feminino , Proteínas do Vírus da Imunodeficiência Humana/administração & dosagem , Proteínas do Vírus da Imunodeficiência Humana/genética , Proteínas do Vírus da Imunodeficiência Humana/imunologia , Imunização/métodos , Memória Imunológica , Camundongos , Plasmídeos , Retinal Desidrogenase/administração & dosagem , Retinal Desidrogenase/genética , Tretinoína/imunologia , Tretinoína/metabolismo , Vacinas de DNA/administração & dosagem , Vacínia/imunologia , Vacínia/prevenção & controle , Vaccinia virus/genéticaRESUMO
Food allergy represents failure to develop tolerance to dietary proteins. Food allergy has increased in prevalence in parallel with decreased exposure to microbes during infancy. In mice, neonatal peroral exposure to the strongly T cell stimulating superantigen staphylococcal enterotoxin A (SEA), enhances the capacity to develop oral tolerance to a novel antigen encountered in adult life. A population of antigen-presenting cells in the gut, the CD103(+) dendritic cells (DCs), is thought to be involved in oral tolerance development, as they convert naïve T cells into FoxP3(+) regulatory T cells (Treg). This function depends on their capacity to convert vitamin A to retinoic acid, carried out by the retinal aldehyde dehydrogenase (RALDH) enzyme. Here, newborn mice were treated with superantigen and DC function and tolerogenic capacity was examined at six weeks of age. We observed that, in mice fed superantigen neonatally, the CD11c(+) DCs had increased expression of RALDH and in vitro more efficiently induced expression Foxp3 expression to stimulated T cells. Further, these mice showed an accumulation of FoxP3(+) T cells in the small intestinal lamina propria and had a more Ag-specific FoxP3(+) T cells after oral tolerance induction in vivo. Moreover, the improved oral tolerance, as shown by increased protection from food allergy, was eradicated if the Vitamin A metabolism was inhibited. These observations contribute to the understanding of how a strong immune stimulation during the neonatal period influences the maturation of the immune system and suggests that such stimulation may reduce the risk of later allergy development.
Assuntos
Animais Recém-Nascidos/imunologia , Antígenos CD/imunologia , Células Dendríticas/imunologia , Enterotoxinas/imunologia , Tolerância Imunológica/imunologia , Cadeias alfa de Integrinas/imunologia , Mucosa Intestinal/imunologia , Superantígenos/imunologia , Animais , Animais Recém-Nascidos/microbiologia , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/microbiologia , Células Dendríticas/microbiologia , Modelos Animais de Doenças , Feminino , Hipersensibilidade Alimentar/imunologia , Hipersensibilidade Alimentar/microbiologia , Fatores de Transcrição Forkhead/imunologia , Mucosa Intestinal/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Ratos , Ratos Sprague-Dawley , Retinal Desidrogenase/imunologia , Staphylococcus aureus/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/microbiologia , Vitamina A/imunologiaRESUMO
Many autoimmune diseases and other chronic inflammatory disorders are characterized by defective FoxP3(+) regulatory T-cell (Treg) mediated suppression. A potential treatment option for these disorders is to increase the number and activity of Tregs locally. Both PLGA (poly-lactic-co-glycolic acid) and TMC-TPP (N-trimethyl chitosan tripolyphosphate) nanoparticles (NP) have been described to enhance T cell activation upon nasal application. Since, PLGA NP and TMC-TPP NP differentially affect CD4(+) T-cell differentiation, we investigated in vitro the capacity of both delivery systems to trigger retinoic acid (RA) production in dendritic cells (DCs) as a strategy to enhance the induction of FoxP3(+) T-cells. We generated ovalbumin (OVA)-encapsulated PLGA NP and TMC-TPP NP that were similar in size (400nm) but differed in their surface charge and other physico-chemical properties. We demonstrate that OVA-specific T-cells that are activated by cervical lymph node (CLN)-derived DCs treated with PLGA NP or TMC-TPP NP show more FoxP3 expression than T-cells that are activated by inguinal lymph node (ILN) cells. We demonstrate that only OVA-encapsulated PLGA NP enhance the induction of FoxP3 in activated T-cells via a TGF-ß and RA dependent mechanism by enhancing retinaldehyde dehydrogenase enzyme (RALDH) expression in CLN-derived DCs that is required for RA production. Additionally, detailed analysis of the CD4(+) T-cell response reveals that PLGA NP induce both IL-10 and IFN-γ production, while TMC-TPP NP induce mainly Th17 production. Underlining that both APC origin and NP characteristics determine the expression level of FoxP3 in activated T-cells. In conclusion, our data suggest that PLGA NP enhance the induction of FoxP3(+) T-cells in the CLN through modulation of DC function and we suggest that they might be a suitable nasal delivery system to treat a wide variety of autoimmune diseases and other chronic inflammatory disorders.
Assuntos
Linfócitos T CD4-Positivos/imunologia , Células Dendríticas/efeitos dos fármacos , Fatores de Transcrição Forkhead/imunologia , Isoenzimas/imunologia , Ácido Láctico/administração & dosagem , Nanopartículas/administração & dosagem , Ácido Poliglicólico/administração & dosagem , Retinal Desidrogenase/imunologia , Família Aldeído Desidrogenase 1 , Animais , Citocinas/imunologia , Células Dendríticas/enzimologia , Células Dendríticas/imunologia , Feminino , Linfonodos/citologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Ovalbumina/imunologia , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Receptores de Antígenos de Linfócitos T/imunologiaRESUMO
PURPOSE: Cancer-initiating cells (CIC) are considered to represent the subpopulation of tumor cells that is resistant to conventional cancer treatments, highly tumorigenic in immunodeficient mice, and responsible for tumor recurrence and metastasis. Based on an elevated aldehyde dehydrogenase (ALDH) activity attributable to ALDH1/3 isoforms, ALDH(bright) cells have been identified and isolated from tumors and shown to have characteristics of CIC. The ALDH1A1 isoform was previously identified as a tumor antigen recognized by CD8(+) T cells. This study examines the ability of ALDH1A1-specific CD8(+) T cells to eliminate ALDH(bright) cells and control tumor growth and metastases. EXPERIMENTAL DESIGN: ALDH(bright) cells were isolated by flow cytometry using ALDEFLUOR from HLA-A2(+) human head and neck, breast, and pancreas carcinoma cell lines and tested for their tumorigenicity in immunodeficient mice. ALDH1A1-specific CD8(+) T cells were generated in vitro and tested for their ability to eliminate CICs in vitro and in vivo by adoptive transfer to immunodeficient mice bearing human tumor xenografts. RESULTS: ALDH(bright) cells isolated by flow cytometry from HLA-A2(+) breast, head and neck, and pancreas carcinoma cell lines at low numbers (500 cells) were tumorigenic in immunodeficient mice. ALDH(bright) cells present in these cell lines, xenografts, or surgically removed lesions were recognized by ALDH1A1-specific CD8(+) T cells in vitro. Adoptive therapy with ALDH1A1-specific CD8(+) T cells eliminated ALDH(bright) cells, inhibited tumor growth and metastases, or prolonged survival of xenograft-bearing immunodeficient mice. CONCLUSIONS: The results of this translational study strongly support the potential of ALDH1A1-based immunotherapy to selectively target CICs in human cancer.