Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
AAPS J ; 23(2): 32, 2021 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-33629139

RESUMO

As one of the most important metabolites of vitamin A, all-trans retinoic acid (RA) plays a crucial role in regulating immune responses. RA has been shown to promote the differentiation of naïve T and B cells and perform diverse functions in the presence of different cytokines. RA also induces gut tropic lymphocytes through upregulating the expression of chemokine (C-C motif) receptor 9 (CCR9) and α4ß7 integrin. In addition, RA promotes the expression of the enzyme retinal dehydrogenase (RALDH) on dendritic cells, which in turn strengthens the ability to synthesize RA. Due to the insolubility of RA, proper formulation design can maximize its ability to improve immune responses for vaccines. Recent studies have developed some formulations co-loading RA and antigen, which can effectively imprint lymphocytes gut homing properties and induce intestine immune responses as well as systemic responses through parenteral administration, providing a promising direction for the protection against mucosal infections. Here, we review the mechanism and effects of RA on lymphocyte differentiation and gut homing, and recent progress of RA delivery systems to improve mucosal immune responses.


Assuntos
Portadores de Fármacos/química , Imunidade nas Mucosas/efeitos dos fármacos , Enteropatias/prevenção & controle , Tretinoína/administração & dosagem , Vacinas/administração & dosagem , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/química , Animais , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/imunologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Modelos Animais de Doenças , Composição de Medicamentos/métodos , Humanos , Imunogenicidade da Vacina , Enteropatias/imunologia , Enteropatias/microbiologia , Mucosa Intestinal/citologia , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/imunologia , Camundongos , Solubilidade , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Tretinoína/química , Tretinoína/imunologia , Excipientes de Vacinas/química , Vacinas/química , Vacinas/imunologia
2.
J Allergy Clin Immunol ; 148(1): 182-194.e4, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33378690

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/imunologia
3.
Biochem Biophys Res Commun ; 516(3): 626-631, 2019 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-31235250

RESUMO

The disruption of intestinal mucosal immune tolerance can lead to the development of intestinal immune diseases such as food allergy (FA). Regulatory T cells (Tregs) in the mucosa play a critical role in maintaining peripheral immune tolerance in the intestine, and retinoic acid (RA) is absolutely required for the induction of Tregs. We have previously reported that kakkonto, a traditional Japanese herbal medicine, suppresses FA in a murine FA model due to the induction of Tregs in the colonic mucosa. However, the precise molecular mechanisms underlying the induction of Tregs remain unclear. Puerarin, an isoflavone derivative, is a major constituent of kakkonto. Thus, we investigated the effect of puerarin on the induction of Tregs. BALB/c mice were systemically sensitized and then orally challenged with ovalbumin (OVA) as an FA model. Puerarin treatment suppressed the development of allergic diarrhea in FA mice. The gene expression levels of IL-4 and mast cell protease I (mMCP-1) were significantly upregulated in the proximal colon of FA mice but were reduced by puerarin. The proportions of Foxp3+CD4+ cells and CD103+CD11c+ dendritic cells (DCs) were significantly higher among the colonic lamina propria (cLP) cells of puerarin-treated FA mice than among those of untreated FA mice. The gene expression of Aldh1a1, an RA synthesis enzyme, in colonic epithelial cells (CECs) was significantly higher in the puerarin-treated FA mouse colon than in the untreated FA mouse colon. In addition, the preventive effect of puerarin was suppressed in the FA model by pretreatment with LE540, an RA receptor (RAR) antagonist. The induction of Foxp3+CD4+ cells and CD103+CD11c+ DCs by puerarin was reduced by pretreatment with LE540. The present findings indicate that the augmentation of RA production in CECs induced by puerarin enhances the induction of Tregs and suppresses the development of FA in a mouse model. Thus, a natural enhancer of RA production, such as puerarin, has the potential to treat immune diseases attributed to Treg deficiency.


Assuntos
Hipersensibilidade Alimentar/imunologia , Tolerância Imunológica/efeitos dos fármacos , Mucosa Intestinal/imunologia , Isoflavonas/farmacologia , Linfócitos T Reguladores/imunologia , Tretinoína/imunologia , Animais , Células Cultivadas , Células Dendríticas/citologia , Células Dendríticas/imunologia , Modelos Animais de Doenças , Fatores de Transcrição Forkhead/imunologia , Fatores de Transcrição Forkhead/metabolismo , Tolerância Imunológica/imunologia , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Transgênicos , Linfócitos T Reguladores/metabolismo , Tretinoína/metabolismo , Vasodilatadores/farmacologia
4.
EMBO Rep ; 20(5)2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30894405

RESUMO

Regulatory T (Treg) cells help to maintain tolerance and prevent the development of autoimmune diseases. Retinoic acid (RA) can promote peripheral conversion of naïve T cells into Foxp3+ Treg cells. Here, we show that RA can act as an adjuvant to induce antigen-specific type 1 Treg (Tr1) cells, which is augmented by co-administration of IL-2. Immunization of mice with the model antigen KLH in the presence of RA and IL-2 induces T cells that secrete IL-10, but not IL-17 or IFN-γ, and express LAG-3, CD49b and PD-1 but not Foxp3, a phenotype typical of Tr1 cells. Furthermore, immunization of mice with the autoantigen MOG in the presence of RA and IL-2 induces Tr1 cells, which suppress pathogenic Th1 and Th17 cells that mediate the development of experimental autoimmune encephalomyelitis (EAE), an autoimmune disease of the CNS. Furthermore, immunization with a surrogate autoantigen, RA and IL-2 prevents development of spontaneous autoimmune uveitis. Our findings demonstrate that the induction of autoantigen-specific Tr1 cells can prevent the development of autoimmunity.


Assuntos
Autoantígenos/imunologia , Autoimunidade/imunologia , Linfócitos T Reguladores/imunologia , Tretinoína/imunologia , Animais , Encefalomielite Autoimune Experimental/imunologia , Feminino , Fatores de Transcrição Forkhead/imunologia , Interleucina-10/imunologia , Interleucina-17/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Células Th1/imunologia , Células Th17/imunologia
5.
J Immunol ; 200(8): 2670-2676, 2018 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-29500243

RESUMO

Many classical vaccines contain whole pathogens and, thus, may occasionally induce adverse effects, such as inflammation. Vaccines containing purified rAgs resolved this problem, but, owing to their low antigenicity, they require adjuvants. Recently, the use of several cytokines, including thymic stromal lymphopoietin (TSLP), has been proposed for this purpose. However, it is difficult to use cytokines as vaccine adjuvants in clinical practice. In this study, we examined the effects of all-trans retinoic acid (atRA) on TSLP production and Ag-induced Ab production. Application of atRA onto the ear lobes of mice selectively induced TSLP production without inducing apparent inflammation. The effects appeared to be regulated via retinoic acid receptors γ and α. Treatment with atRA was observed to enhance OVA-induced specific Ab production; however, this effect was completely absent in TSLP receptor-knockout mice. An enhancement in Ab production was also observed when recombinant hemagglutinin was used as the Ag. In conclusion, atRA was an effective adjuvant through induction of TSLP production. Therefore, we propose that TSLP-inducing low m.w. compounds, such as atRA, may serve as effective adjuvants for next-generation vaccines.


Assuntos
Formação de Anticorpos/imunologia , Citocinas/imunologia , Tretinoína/imunologia , Animais , Antígenos/imunologia , Feminino , Hemaglutininas/imunologia , Inflamação/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Knockout , Linfopoietina do Estroma do Timo
6.
Vaccine ; 35(28): 3534-3547, 2017 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-28545924

RESUMO

Bacterial outer membrane vesicles have been extensively investigated and considered as a next generation vaccine. Recently, we have demonstrated that the cholera pentavalent outer membrane vesicles (CPMVs) immunogen induced adaptive immunity and had a strong protective efficacy against the circulating V. cholerae strains in a mouse model. In this present study, we are mainly focusing on reducing outer membrane vesicle (OMV) -mediated toxicity without altering its antigenic property. Therefore, we have selected All-trans Retinoic Acid (ATRA), active metabolites of vitamin A, which have both anti-inflammatory and mucosal adjuvant properties. Pre-treatment of ATRA significantly reduced CPMVs induced TLR2 mediated pro-inflammatory responses in vitro and in vivo. Furthermore, we also found ATRA pre-treatment significantly induced mucosal immune response and protective efficacy after two doses of oral immunization with CPMVs (75µg). This study can help to reduce OMV based vaccine toxicity and induce better protective immunity where children and men suffered from malnutrition mainly in developing countries.


Assuntos
Proteínas da Membrana Bacteriana Externa/imunologia , Vacinas contra Cólera/imunologia , Imunidade nas Mucosas , Inflamação/prevenção & controle , Tretinoína/administração & dosagem , Vibrio cholerae/imunologia , Administração Oral , Animais , Animais Recém-Nascidos , Anticorpos Antibacterianos/sangue , Proteínas da Membrana Bacteriana Externa/toxicidade , Cólera/imunologia , Cólera/prevenção & controle , Vacinas contra Cólera/administração & dosagem , Vacinas contra Cólera/toxicidade , Citocinas/biossíntese , Modelos Animais de Doenças , Feminino , Imunogenicidade da Vacina , Imunoglobulina G/sangue , Camundongos , Camundongos Endogâmicos BALB C , Receptor 2 Toll-Like/imunologia , Tretinoína/imunologia , Tretinoína/farmacologia
7.
Vaccine ; 34(46): 5629-5635, 2016 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-27670072

RESUMO

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/genética
8.
Eur J Immunol ; 45(5): 1287-95, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25808452

RESUMO

The importance of vitamin A for host defense is undeniable and the study of its mechanisms is paramount. Of the estimated 250 million preschool children who are vitamin A-deficient (VAD), 10% will die from their increased susceptibility to infectious disease. Vitamin A supplementation was established in the 1980s as one of the most successful interventions in the developing world. Understanding how vitamin A controls immunity will help curb the mortality and morbidity associated with vitamin A deficiency and exploit the immune-enhancing capacity of vitamin A to heighten host resistance to infectious disease. The discoveries that retinoic acid (RA) imprints the homing of leukocytes to the gut and enhances the induction of regulatory T cells, highlighted a potential role for RA in mucosal tolerance. However, more recently emerging data tell of a more profound systemic impact of RA on leukocyte function and commitment. In animal models using genetic manipulation of RA signaling, we learned when and how RA controls T cell fate. Here, we review the role for RA as a critical checkpoint regulator in the differentiation of CD4(+) T cells within the immune system.


Assuntos
Vitamina A/imunologia , Animais , Diferenciação Celular , Fatores de Transcrição Forkhead/metabolismo , Humanos , Imunidade nas Mucosas , Imunossupressores/uso terapêutico , Imunoterapia , Camundongos , Modelos Imunológicos , Retinoides/uso terapêutico , Transdução de Sinais , Linfócitos T Auxiliares-Indutores/citologia , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/metabolismo , Tretinoína/imunologia , Tretinoína/metabolismo , Deficiência de Vitamina A/imunologia
9.
Vaccine ; 32(27): 3386-92, 2014 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-24780251

RESUMO

Vaccination is an important tool for enhancing immune responses against mucosal pathogens. Intramuscularly administered adenovirus (Ad) vectors have been demonstrated to be strong inducers of both systemic and mucosal immune responses. Further enhancement of immune responses following Ad vaccination is highly desirable. All-trans retinoic acid (ATRA), a biologically active vitamin A metabolite, has been explored as an adjuvant for primary immune responses following vaccination. In this study, we investigated the effect of ATRA on a heterologous Ad prime boost regimen. ATRA co-administration during priming increased mucosal and systemic antibody responses as well as mucosal but not systemic CD8(+) T cell responses. However, this effect was no longer apparent after boosting regardless of whether ATRA was administered at the time of priming, at the time of boosting, or at both immunizations. Our findings confirm ATRA as an adjuvant for primary immune responses and suggest that the adjuvant effect does not extend to secondary immune responses.


Assuntos
Vacinas contra a AIDS/imunologia , Linfócitos T CD8-Positivos/imunologia , Vetores Genéticos/imunologia , Imunidade nas Mucosas , Tretinoína/imunologia , Adenoviridae/imunologia , Adjuvantes Imunológicos/administração & dosagem , Animais , Feminino , Imunização Secundária , Camundongos Endogâmicos BALB C , Receptores CCR/metabolismo , Baço/imunologia , Vacinas Sintéticas/imunologia , Produtos do Gene gag do Vírus da Imunodeficiência Humana/imunologia
10.
Clin Exp Immunol ; 175(3): 468-75, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24237035

RESUMO

There is an urgent need to identify ways of enhancing the mucosal immune response to oral vaccines. Rotavirus vaccine protection is much lower in Africa and Asia than in industrialized countries, and no oral vaccine has efficacy approaching the best systemic vaccines. All-trans retinoic acid (ATRA) up-regulates expression of α4ß7 integrin and CCR9 on lymphocytes in laboratory animals, increasing their gut tropism. The aim of this study was to establish the feasibility of using ATRA as an oral adjuvant for oral typhoid vaccination. In order to establish that standard doses of oral ATRA can achieve serum concentrations greater than 10 nmol/l, we measured ATRA, 9-cis and 13-cis retinoic acid in serum of 14 male volunteers before and 3 h after 10 mg ATRA. We then evaluated the effect of 10 mg ATRA given 1 h before, and for 7 days following, oral typhoid vaccine in eight men, and in 24 men given various control interventions. We measured immunoglobulin (Ig)A directed against lipopolysaccharide (LPS)and protein preparations of vaccine antigens in whole gut lavage fluid (WGLF) and both IgA and IgG in serum, 1 day prior to vaccination and on day 14. Median [interquartile range (IQR)] C(max) was 26·2 (11·7-39·5) nmol/l, with no evidence of cumulation over 8 days. No adverse events were observed. Specific IgA responses to LPS (P = 0·02) and protein (P = 0·04) were enhanced in WGLF, but no effect was seen on IgA or IgG in serum. ATRA was well absorbed, well tolerated and may be a promising candidate oral adjuvant.


Assuntos
Adjuvantes Imunológicos , Imunidade nas Mucosas/imunologia , Tretinoína/imunologia , Vacinas Tíficas-Paratíficas/imunologia , Administração Oral , Adulto , Anticorpos Antibacterianos/imunologia , Humanos , Imunoglobulina A Secretora/imunologia , Mucosa Intestinal/imunologia , Masculino , Pessoa de Meia-Idade , Tretinoína/administração & dosagem , Tretinoína/farmacocinética , Vacinas Tíficas-Paratíficas/administração & dosagem , Adulto Jovem , Zâmbia
11.
Immunobiology ; 218(12): 1477-87, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23816303

RESUMO

Mature naïve B cells possess a number of BCR coreceptors and other antigen receptors, including the MHC class I-like molecule CD1d, but little is known of the response of B cells to stimulation by the CD1d ligand, α-galactosylceramide (αGalCer). Previously, we showed that all-trans-retinoic acid (RA) increases the expression of CD1d and the magnitude of CD1d-mediated antibody production in vivo. Potential mechanisms could include changes in the expression of costimulatory molecules and transcription factors that regulate plasma cell formation. In the present study, we have used isolated purified B cells and in vivo studies to demonstrate that αGalCer and RA initiate a regulated expression of several genes essential for B cell activation and differentiation, such as Pax-5, Blimp-1, IRF-4 and activation-induced cytidine deaminase (Aid). Moreover, whereas αGalCer mainly increased the expression of Pax-5, CD40 and CD86 that are critical for B cell activation, RA predominantly increased CD138⁺ and Fas⁺-PNA⁺ B cells, which represent more advanced B cell differentiation. It is also noteworthy that αGalCer enriched a CD19hi subset of B cells, which represent B cells with more differentiated phenotype and higher potential for antibody production. In vivo, treatment with αGalCer enriched the CD19hi population, which, after sorting, produced more anti-TT IgG by ELISPOT assay. Together, our data demonstrate that RA and αGalCer can regulate B cell activation and differentiation at multiple levels in a complementary manner, facilitating the progress of B cells towards antibody secreting cells.


Assuntos
Subpopulações de Linfócitos B/imunologia , Linfócitos B/imunologia , Galactosilceramidas/imunologia , Tretinoína/imunologia , Animais , Formação de Anticorpos , Antígenos CD/metabolismo , Antígenos CD1d/metabolismo , Diferenciação Celular , Células Cultivadas , Citidina Desaminase/metabolismo , Feminino , Galactosilceramidas/agonistas , Fatores Reguladores de Interferon/metabolismo , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Fator de Transcrição PAX5/metabolismo , Fator 1 de Ligação ao Domínio I Regulador Positivo , Receptores de Antígenos de Linfócitos B/metabolismo , Toxoide Tetânico/imunologia , Fatores de Transcrição/metabolismo
12.
J Innate Immun ; 4(2): 176-86, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22213773

RESUMO

Patients with vitamin A/retinol deficiency are shown to be prone to infections and to suffer from increased inflammation, effects which can be remedied by vitamin A supplements. We aimed to study how human monocytes from the peripheral venous blood of healthy donors acted within the initial hours after adherence and exposure to bacterial endotoxin in the presence or absence of the 9-cis-isomer of retinoic acid (9cisRA). We found that adherent human monocytes were dominated by the CD14dimCD16+ subtype. Pretreatment with 9cisRA for 1 h significantly decreased lipopolysaccharide (LPS)-induced mRNA expression and protein release of tumor necrosis factor (TNF)α, interleukin (IL)-6 and chemokine ligands (CCL)3 and CCL4. In contrast, treatment with 9cisRA rapidly enhanced the production of monocyte chemoattractive protein/CCL2. 9cisRA treatment also led to enhanced migration of classical CD14high monocytes in a transwell in vitro system. We conclude that 9cisRA treatment of human adherent monocytes attenuates the inflammatory responses to LPS and induces the attraction of classical monocytes, a feature which may help explain why supplements administered to vitamin A-deficient patients counteract inflammation and increases the ability to fight infections.


Assuntos
Movimento Celular/imunologia , Inflamação/imunologia , Monócitos/imunologia , Tretinoína/imunologia , Adesão Celular/imunologia , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Citocinas/biossíntese , Citocinas/imunologia , Citometria de Fluxo , Humanos , Inflamação/metabolismo , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tretinoína/metabolismo , Tretinoína/farmacologia
13.
Neurosci Lett ; 509(1): 64-8, 2012 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-22230896

RESUMO

In order to increase our knowledge about the distribution of vitamins in the mammalian brain, we have developed a highly specific antiserum directed against retinoic acid with good affinity (10(-8) M), as evaluated by ELISA tests. In the rat brain, no immunoreactive fibers containing retinoic acid were detected. Cell bodies containing retinoic acid were only found in the hypothalamus. This work reports the first visualization and the morphological characteristics of cell bodies containing retinoic acid in the mammalian paraventricular hypothalamic nucleus and in the dorsal perifornical region, using an indirect immunoperoxidase technique. The restricted distribution of retinoic acid in the rat brain suggests that this vitamin could be involved in very specific physiological mechanisms.


Assuntos
Hipotálamo/química , Tretinoína/análise , Animais , Ensaio de Imunoadsorção Enzimática , Hipotálamo/citologia , Soros Imunes/imunologia , Técnicas Imunoenzimáticas , Imuno-Histoquímica , Núcleo Hipotalâmico Paraventricular/química , Núcleo Hipotalâmico Paraventricular/citologia , Ratos , Tretinoína/imunologia
14.
Clin Vaccine Immunol ; 18(6): 1015-20, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21471300

RESUMO

All-trans-retinoic acid (RA) promotes the maturation and differentiation of B cells, which are known as a type of professional antigen-presenting cells. We show here that CD1d, a major histocompatibility complex class I-like molecule that presents lipid antigens, is expressed in the mouse spleen B cells and is increased by RA. Thus, we hypothesized that RA and the CD1d ligand, α-galactosylceramide (αGalCer), could interact to promote the differentiation, maturation, and antibody response of antigen-activated B cells. In isolated B cells, αGalCer alone markedly stimulated, and RA further increased B cell proliferation, synergizing with the B cell antigen receptor ligation via anti-µ antibody (P < 0.05). The significantly increased cell proliferation stimulated by αGalCer was abrogated in the B cells of CD1d-null mice. RA alone and combined with αGalCer also promoted B cell differentiation by the enrichment of sIgG1-, CD138-, and PNA/Fas-positive B cells (P < 0.05), suggesting a plasmacytic cell differentiation. In vivo, wild-type mice treated with RA and/or αGalCer during primary immunization with tetanus toxoid produced a higher serum anti-tetanus IgG response and had more bone marrow anti-tetanus antibody-secreting cells as determined by enzyme-linked immunospot assay (P < 0.05) in the secondary response, a finding indicative of heightened long-term memory; however, the increased antibody secretion after αGalCer treatment was abolished in CD1d-null mice. We provide evidence here that RA, together with αGalCer, can effectively regulate B cell proliferation and differentiation, ultimately promoting a more efficient antibody response to protein antigen. The results suggest that the combination of RA and αGalCer could be a useful adjuvant combination in vaccine strategies.


Assuntos
Formação de Anticorpos , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Galactosilceramidas/imunologia , Ativação Linfocitária , Tretinoína/imunologia , Animais , Anticorpos Antibacterianos/sangue , Antígenos CD1d/biossíntese , Antígenos CD1d/genética , Antitoxinas/sangue , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Feminino , Imunoglobulina G/sangue , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Toxoide Tetânico/administração & dosagem , Toxoide Tetânico/imunologia
16.
Nature ; 471(7337): 220-4, 2011 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-21307853

RESUMO

Under physiological conditions the gut-associated lymphoid tissues not only prevent the induction of a local inflammatory immune response, but also induce systemic tolerance to fed antigens. A notable exception is coeliac disease, where genetically susceptible individuals expressing human leukocyte antigen (HLA) HLA-DQ2 or HLA-DQ8 molecules develop inflammatory T-cell and antibody responses against dietary gluten, a protein present in wheat. The mechanisms underlying this dysregulated mucosal immune response to a soluble antigen have not been identified. Retinoic acid, a metabolite of vitamin A, has been shown to have a critical role in the induction of intestinal regulatory responses. Here we find in mice that in conjunction with IL-15, a cytokine greatly upregulated in the gut of coeliac disease patients, retinoic acid rapidly activates dendritic cells to induce JNK (also known as MAPK8) phosphorylation and release the proinflammatory cytokines IL-12p70 and IL-23. As a result, in a stressed intestinal environment, retinoic acid acted as an adjuvant that promoted rather than prevented inflammatory cellular and humoral responses to fed antigen. Altogether, these findings reveal an unexpected role for retinoic acid and IL-15 in the abrogation of tolerance to dietary antigens.


Assuntos
Adjuvantes Imunológicos/farmacologia , Doença Celíaca/imunologia , Glutens/imunologia , Interleucina-15/imunologia , Tretinoína/farmacologia , Administração Oral , Adolescente , Adulto , Animais , Doença Celíaca/induzido quimicamente , Doença Celíaca/etiologia , Células Cultivadas , Criança , Pré-Escolar , Técnicas de Cocultura , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/enzimologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Dieta , Fatores de Transcrição Forkhead/metabolismo , Gliadina/administração & dosagem , Gliadina/imunologia , Glutens/administração & dosagem , Antígenos HLA-DQ/genética , Antígenos HLA-DQ/imunologia , Humanos , Tolerância Imunológica/efeitos dos fármacos , Inflamação/imunologia , Interleucina-12/biossíntese , Interleucina-12/imunologia , Interleucina-12/metabolismo , Interleucina-15/genética , Interleucina-23/imunologia , Interleucina-23/metabolismo , Mucosa Intestinal/citologia , Mucosa Intestinal/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pessoa de Meia-Idade , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Fosforilação/efeitos dos fármacos , Receptores de Interleucina-12/deficiência , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Tretinoína/imunologia , Adulto Jovem
17.
Arch Virol ; 156(2): 183-202, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21170730

RESUMO

Modern adjuvants should induce strong and balanced immune responses, and it is often desirable to induce specific types of immunity. As an example, efficient Th1-immunity-inducing adjuvants are highly in demand. Such adjuvants promote good cell-mediated immunity against subunit vaccines that have low immunogenicity themselves. The development of such adjuvants may take advantage of the increased knowledge of the molecular mechanisms and factors controlling these responses. However, knowledge of such molecular details of immune mechanisms is relatively scarce for species other than humans and laboratory rodents, and in addition, there are special considerations pertaining to the use of adjuvants in veterinary animals, such as production and companion animals. With a focus on veterinary animals, this review highlights a number of approaches being pursued, including cytokines, CpG oligonucleotides, microparticles and liposomes.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Vacinação/veterinária , Vacinas/administração & dosagem , Imunidade Adaptativa , Adjuvantes Imunológicos/efeitos adversos , Animais , Animais Domésticos/imunologia , Sistemas de Liberação de Medicamentos/métodos , Sistemas de Liberação de Medicamentos/tendências , Sistemas de Liberação de Medicamentos/veterinária , Imunidade Inata , Imunidade nas Mucosas , Interferons/administração & dosagem , Interferons/imunologia , Lipossomos , Microesferas , Neoplasias/etiologia , Neoplasias/veterinária , Oligodesoxirribonucleotídeos/administração & dosagem , Oligodesoxirribonucleotídeos/imunologia , Receptores Toll-Like/agonistas , Receptores Toll-Like/imunologia , Tretinoína/administração & dosagem , Tretinoína/imunologia , Vacinação/métodos , Vacinação/tendências , Vacinas/efeitos adversos
18.
Br J Ophthalmol ; 94(6): 802-7, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19965837

RESUMO

AIMS: To determine whether an active metabolite of vitamin A, all-trans retinoic acid (ATRA), reduces inflammation in experimental autoimmune uveoretinitis (EAU). METHODS: Naive CD4(+) T cells were activated with anti-CD3, anti-CD28 and transforming growth factor (TGF)-beta, in the presence or absence of ATRA. Intracellular expression of transcription factor forkhead box P3 (Foxp3) and interleukin (IL)-17 in the activated CD4(+) T cells was assessed by flow cytometry. C57BL/6 mice were immunised with human interphotoreceptor retinoid binding protein peptide 1-20 (IRBP(1-20)). ATRA was administered intraperitoneally every other day (0.2 mg/mouse per day) from day 0 to day 21. In vivo-primed draining lymph node cells from vehicle-treated or ATRA-treated mice were stimulated with IRBP(1-20) and the culture supernatant fraction was harvested for assay of interferon (IFN)-gamma and IL-17 by ELISA. RESULTS: ATRA synergised with TGF-beta to induce Foxp3(+) T regulatory cells (Treg) and reciprocally inhibited development of IL-17-producing T helper cells (Th17) induced by TGF-beta and IL-6. ATRA treatment reduced the severity of EAU clinically, and IFN-gamma and IL-17 production were significantly reduced in ATRA-treated mice. CONCLUSION: These findings demonstrate that ATRA treatment ameliorates severity of EAU and reduces the Th1/Th17 responses. ATRA may represent a new therapeutic modality for human refractory uveitis.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Doenças Autoimunes/tratamento farmacológico , Retinite/tratamento farmacológico , Tretinoína/uso terapêutico , Uveíte/tratamento farmacológico , Animais , Anti-Inflamatórios não Esteroides/imunologia , Doenças Autoimunes/imunologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos/métodos , Feminino , Fatores de Transcrição Forkhead/análise , Interferon gama/biossíntese , Interleucina-17/biossíntese , Linfonodos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Interleucina-6/metabolismo , Retinite/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Tretinoína/imunologia , Uveíte/imunologia
19.
Transplant Proc ; 41(6): 2670-2, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19715998

RESUMO

CD4(+)CD25(+)Foxp3(+) regulatory T cells (Treg) mediate immunologic self-tolerance and suppress immune responses. In the gut, a subset of dendritic cells is specialized to induce Treg in a transforming growth factor-beta (TGF-beta)- and retinoic acid (RA)-dependent manner. The aim of this study was to establish if RA synergizing with TGF-beta induced antigen specific CD4(+) CD25(high) Foxp3(+) Treg portraying gut homing receptors. Splenic CD4(+)CD25(-) Foxp3(-) naïve T cells from DO11.10 mice were cocultured with splenic CD11c(+) dendritic cells from Balb/c mice in the presence of TGF-beta, RA, and low levels of an antigenic peptide. After 5 days of culture, cells were analyzed for the expression of Foxp3 and the gut homing receptors CCR9 and alpha4beta7. The number of Foxp3(+) T cells generated with TGF-beta and RA was at least 3 times higher than in the cultures with TGF-beta alone and 15 times higher than in controls without exogenous cytokines. Also, supplementation of the cultures with RA induced the expression of the intestinal homing receptors CCR9 and alpha4beta7. Our results showed that coculture of naïve T cells with antigen-presenting cells in the presence of TGF-beta and RA represents a powerful approach to generate Treg with specific homing receptors.


Assuntos
Receptores de Retorno de Linfócitos/genética , Linfócitos T Reguladores/imunologia , Fator de Crescimento Transformador beta/imunologia , Tretinoína/imunologia , Animais , Antígeno CD11c/imunologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Diferenciação Celular , Células Cultivadas , Feminino , Citometria de Fluxo , Fatores de Transcrição Forkhead/imunologia , Subunidade alfa de Receptor de Interleucina-2/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos , Tolerância a Antígenos Próprios/imunologia , Baço/citologia , Baço/efeitos dos fármacos , Baço/imunologia , Tretinoína/farmacologia
20.
Vitam Horm ; 75: 197-222, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17368317

RESUMO

Vitamin A (VA, retinol) is essential for normal immune system maturation, but the effect of VA(1) on antibody production, the hallmark of successful vaccination, is still not well understood. In countries where VA deficiency is a public health problem, many children worldwide are now receiving VA along with immunizations against poliovirus, measles, diphtheria, pertussis, and tetanus. The primary goal has been to provide enough VA to protect against the development of VA deficiency for a period of 4-6 months. However, it is also possible that VA might promote the vaccine antibody response. Several community studies, generally of small size, have been conducted in children supplemented with VA at the time of immunization, as promoted by the World Health Organization/UNICEF. However, only a few studies have reported differences in antibody titers or seroconversion rates due to VA. However, VA status was not directly assessed, and in some communities children were often breast fed, another strategy for preventing VA deficiency. Some of the vaccines used induced a high rate of seroconversion, even without VA. In children likely to have been VA deficient, oral polio vaccine seroconversion rate was increased by VA. In animal models, where VA status was controlled and VA deficiency confirmed, the antibody response to T-cell-dependent (TD) and polysaccharide antigens was significantly reduced, congruent with other defects in innate and adaptive immunity. Moreover, the active metabolite of VA, retinoic acid (RA) can potentiate antibody production to TD antigens in normal adult and neonatal animals. We speculate that numerous animal studies have correctly identified VA deficiency as a risk factor for low antibody production. A lack of effect of VA in human studies could be due to a low rate of VA deficiency in the populations studied or low sample numbers. The ability to detect differences in antibody response may also depend on the vaccine-adjuvant combination used. Future studies of VA supplementation and immunization should include assessment of VA status and a sufficiently large sample size. It would also be worthwhile to test the effect of neonatal VA supplementation on the response to immunization given after 6 months to 1 year of age, as VA supplementation, by preventing the onset of VA deficiency, may improve the response to immunizations given later on.


Assuntos
Formação de Anticorpos/efeitos dos fármacos , Suplementos Nutricionais , Tretinoína/uso terapêutico , Vitamina A/uso terapêutico , Vitaminas/uso terapêutico , Animais , Formação de Anticorpos/imunologia , Humanos , Vacinação em Massa/métodos , Tretinoína/imunologia , Vitamina A/administração & dosagem , Vitamina A/imunologia , Deficiência de Vitamina A/imunologia , Deficiência de Vitamina A/prevenção & controle , Vitaminas/administração & dosagem , Vitaminas/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA