Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
J Immunol ; 197(6): 2239-49, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27527592

RESUMEN

Human and mouse neonates exhibit limited vaccine responses characterized by predominant Th2 and limited Th1 responses. Because IL-36 exerts a synergic adjuvant effect with IL-12, enhancing Th1 polarization in adult (AD) mice, we administered IL-36ß to neonatal (1-wk old) and AD control mice at the time of immunization with tetanus toxoid adsorbed to aluminum hydroxide (TT/Alum). Unexpectedly, the combination of IL-36ß with TT/Alum, which was well tolerated in AD mice, proved toxic and even lethal in neonates. This neonatal toxicity was associated with high Il36r mRNA expression in neonatal liver, resulting in increased cytokine production. Liver Il36r mRNA expression decreased with the termination of fetal liver hematopoiesis, and this decrease correlated with a complete protection from TT/Alum/IL-36ß-induced mortality. The combination of IL-36ß and TT/Alum induced the rapid production of TNF-α and IFN-γ by liver myeloid and lymphoid cells, respectively. These responses were less marked when IL-36ß was used alone, with no adverse effect. The toxicity of IL-36ß + TT/Alum was abrogated by the administration of a neutralizing anti-TNF-α Ab, confirming causality. In conclusion, liver myeloid cells in neonatal mice are an important source of proinflammatory cytokines that may lead to TNF-α-mediated toxicity and even lethality.


Asunto(s)
Interleucina-1/toxicidad , Hígado/inmunología , Células Mieloides/inmunología , Factor de Necrosis Tumoral alfa/biosíntesis , Animales , Animales Recién Nacidos , Citocinas/biosíntesis , Ratones , Ratones Endogámicos C57BL , Toxoide Tetánico/toxicidad
2.
PLoS Pathog ; 11(3): e1004715, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25763578

RESUMEN

Interleukin-21 signaling is important for germinal center B-cell responses, isotype switching and generation of memory B cells. However, a role for IL-21 in antibody-mediated protection against pathogens has not been demonstrated. Here we show that IL-21 is produced by T follicular helper cells and co-expressed with IFN-γ during an erythrocytic-stage malaria infection of Plasmodium chabaudi in mice. Mice deficient either in IL-21 or the IL-21 receptor fail to resolve the chronic phase of P. chabaudi infection and P. yoelii infection resulting in sustained high parasitemias, and are not immune to re-infection. This is associated with abrogated P. chabaudi-specific IgG responses, including memory B cells. Mixed bone marrow chimeric mice, with T cells carrying a targeted disruption of the Il21 gene, or B cells with a targeted disruption of the Il21r gene, demonstrate that IL-21 from T cells signaling through the IL-21 receptor on B cells is necessary to control chronic P. chabaudi infection. Our data uncover a mechanism by which CD4+ T cells and B cells control parasitemia during chronic erythrocytic-stage malaria through a single gene, Il21, and demonstrate the importance of this cytokine in the control of pathogens by humoral immune responses. These data are highly pertinent for designing malaria vaccines requiring long-lasting protective B-cell responses.


Asunto(s)
Linfocitos B/inmunología , Inmunidad Humoral/inmunología , Interleucinas/inmunología , Malaria/inmunología , Transducción de Señal , Linfocitos T/inmunología , Animales , Comunicación Celular/inmunología , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Plasmodium chabaudi/inmunología , Plasmodium yoelii/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa
3.
J Immunol ; 194(10): 4836-45, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25870238

RESUMEN

The early life influenza disease burden calls for more effective vaccines to protect this vulnerable population. Influenza vaccines including the MF59 oil-in-water adjuvant induce higher, broader, and more persistent Ab responses in adults and particularly in young, through yet undefined mechanisms. In this study, we show that MF59 enhances adult murine IgG responses to influenza hemagglutinin (HA) by promoting a potent T follicular helper cells (TFH) response, which directly controls the magnitude of the germinal center (GC) B cell response. Remarkably, this enhancement of TFH and GC B cells is already fully functional in 3-wk-old infant mice, which were fully protected by HA/MF59 but not HA/PBS immunization against intranasal challenge with the homologous H1N1 (A/California/7/2009) strain. In 1-wk-old neonatal mice, MF59 recruits and activates APCs, efficiently induces CD4(+) effector T cells and primes for enhanced infant responses but induces few fully functional TFH cells, which are mostly follicular regulatory T cells, and poor GC and anti-HA responses. The B cell adjuvanticity of MF59 appears to be mediated by the potent induction of TFH cells which directly controls GC responses both in adult and early life, calling for studies assessing its capacity to enhance the efficacy of influenza immunization in young infants.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Vacunas contra la Influenza/inmunología , Polisorbatos/farmacología , Escualeno/farmacología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Animales Recién Nacidos , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Centro Germinal/inmunología , Inmunohistoquímica , Subtipo H1N1 del Virus de la Influenza A/inmunología , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Escualeno/inmunología
4.
Front Immunol ; 14: 1119371, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36845155

RESUMEN

Background: The use of circulating cDC1 to generate anti-cancer vaccines is among the most promising approaches to overcome the limited immunogenicity and clinical efficacy of monocyte-derived DC. However, the recurrent lymphopenia and the reduction of DC numbers and functionality in patients with cancer may represent an important limitation of such approach. In patients with ovarian cancer (OvC) that had received chemotherapy, we previously showed that cDC1 frequency and function were reduced. Methods: We recruited healthy donors (HD, n=7) and patients with OvC at diagnosis and undergoing interval debulking surgery (IDS, n=6), primary debulking surgery (PDS, n=6) or at relapse (n=8). We characterized longitudinally phenotypic and functional properties of peripheral DC subsets by multiparametric flow cytometry. Results: We show that the frequency of cDC1 and the total CD141+ DC capacity to take up antigen are not reduced at the diagnosis, while their TLR3 responsiveness is partially impaired in comparison with HD. Chemotherapy causes cDC1 depletion and increase in cDC2 frequency, but mainly in patients belonging to the PDS group, while in the IDS group both total lymphocytes and cDC1 are preserved. The capacity of total CD141+ DC and cDC2 to take up antigen is not impacted by chemotherapy, while the activation capacity upon Poly(I:C) (TLR3L) stimulation is further decreased. Conclusions: Our study provides new information about the impact of chemotherapy on the immune system of patients with OvC and sheds a new light on the importance of considering timing with respect to chemotherapy when designing new vaccination strategies that aim at withdrawing or targeting specific DC subsets.


Asunto(s)
Células Dendríticas , Recurrencia Local de Neoplasia , Neoplasias Ováricas , Femenino , Humanos , Inmunoterapia , Monocitos , Neoplasias Ováricas/tratamiento farmacológico , Células Dendríticas/inmunología
5.
Front Immunol ; 14: 1155200, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37063899

RESUMEN

Introduction: C-type lectin receptor (CLR) agonists emerged as superior inducers of primary B cell responses in early life compared with Toll-like receptor (TLR) agonists, while both types of adjuvants are potent in adults. Methods: Here, we explored the mechanisms accounting for the differences in neonatal adjuvanticity between a CLR-based (CAF®01) and a TLR4-based (GLA-SE) adjuvant administered with influenza hemagglutinin (HA) in neonatal mice, by using transcriptomics and systems biology analyses. Results: On day 7 after immunization, HA/CAF01 increased IL6 and IL21 levels in the draining lymph nodes, while HA/GLA-SE increased IL10. CAF01 induced mixed Th1/Th17 neonatal responses while T cell responses induced by GLA-SE had a more pronounced Th2-profile. Only CAF01 induced T follicular helper (Tfh) cells expressing high levels of IL21 similar to levels induced in adult mice, which is essential for germinal center (GC) formation. Accordingly, only CAF01- induced neonatal Tfh cells activated adoptively transferred hen egg lysozyme (HEL)-specific B cells to form HEL+ GC B cells in neonatal mice upon vaccination with HEL-OVA. Discussion: Collectively, the data show that CLR-based adjuvants are promising neonatal and infant adjuvants due to their ability to harness Tfh responses in early life.


Asunto(s)
Linfocitos B , Centro Germinal , Lectinas Tipo C , Células T Auxiliares Foliculares , Animales , Ratones , Adyuvantes Inmunológicos/farmacología , Lectinas Tipo C/agonistas , Animales Recién Nacidos
6.
Cancers (Basel) ; 15(12)2023 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-37370724

RESUMEN

Benign prostate hyperplasia (BPH) is a frequent condition in aging men, which affects life quality, causing principally lower urinary tract symptoms. Epidemiologic studies suggest that BPH may raise the risk of developing prostate cancer (PCa), most likely promoting a chronic inflammatory environment. Studies aiming at elucidating the link and risk factors that connect BPH and PCa are urgently needed to develop prevention strategies. The BPH microenvironment, similar to the PCa one, increases immune infiltration of the prostate, but, in contrast to PCa, immunosuppression may not be established yet. In this study, we found that prostate-infiltrating lymphocytes (PILs) expanded from hyperplastic prostate tissue recognized tumor-associated antigens (TAA) and autologous tissue, regardless of the presence of tumor cells. PILs expanded from BPH samples of patients with PCa, however, seem to respond more strongly to autologous tissue. Phenotypic characterization of the infiltrating PILs revealed a trend towards better expanding CD4+ T cells in infiltrates derived from PCa, but no significant differences were found. These findings suggest that T cell tolerance is compromised in BPH-affected prostates, likely due to qualitative or quantitative alterations of the antigenic landscape. Our data support the hypothesis that BPH increases the risk of PCa and may pave the way for new personalized preventive vaccine strategies for these patients.

7.
Front Immunol ; 11: 580974, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33262759

RESUMEN

Novel adjuvants, such as Toll-like receptors (TLRs) agonists, are needed for the development of new formulations able to circumvent limitations of current vaccines. Among TLRs, TLR7/8 agonists represent promising candidates, as they are well described to enhance antigen-specific antibody responses and skew immunity toward T helper (TH) 1 responses. We find here that the incorporation of the synthetic TLR7/8 ligand 3M-052 in a cationic DOEPC-based liposome formulation shifts immunity toward TH1 responses and elicits strong and long-lasting germinal center and follicular T helper cell responses in adult mice. This reflects the prolonged recruitment of innate cells toward the site of immunization and homing of activated antigen-loaded monocytes and monocyte-derived dendritic cells toward draining lymph nodes. We further show that this adjuvanticity is independent of type I IFN but NF-κB-dependent. Overall, our data identify TLR7/8 agonists incorporated in liposomes as promising and effective adjuvants to enhance TH1 and germinal center responses.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Glicoproteínas de Membrana/agonistas , Monocitos/inmunología , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 8/agonistas , Animales , Linfocitos B/inmunología , Células Dendríticas/inmunología , Composición de Medicamentos , Centro Germinal/inmunología , Compuestos Heterocíclicos con 3 Anillos/administración & dosificación , Inmunidad Innata , Interferón Tipo I/inmunología , Ligandos , Liposomas/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/deficiencia , FN-kappa B/genética , FN-kappa B/inmunología , Fosfatidilcolinas/administración & dosificación , Receptor de Interferón alfa y beta/deficiencia , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/inmunología , Transducción de Señal/inmunología , Ácidos Esteáricos/administración & dosificación , Células TH1/inmunología
8.
Cancer Immunol Res ; 8(4): 556-564, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32019778

RESUMEN

Innate lymphoid cells (ILC) are a family of immune cells that are emerging as potent orchestrators of immune responses. In cancer, ILCs display both pro- and antitumorigenic functions depending on the nature of the tumor and the involved ILC subset. Little is known about the ILC-tumor cross-talk in human melanoma. Here, we showed that ILC1s were enriched but functionally impaired in cytokine secretion in both peripheral blood mononuclear cells and tumor-infiltrated lymph nodes of melanoma patients. These findings were confirmed in vivo in murine cutaneous melanoma. Multiple immunosuppressive mechanisms are described in the melanoma microenvironment. Among others, adenosine and kynurenines were shown to suppress antitumor immune responses. By exposing ILCs to adenosine and kynurenines, we observed a similar shift toward the ILC1 subset distribution and impairment in proinflammatory cytokine production to that of patient samples studied ex vivo. Thus, we hypothesized that the immunosuppressive microenvironment of malignant melanoma might shape ILC subpopulations. Hence, we provide a rationale for the use of drugs targeting adenosine and kynurenine pathways in melanoma patients.


Asunto(s)
Inmunidad Innata , Inmunosupresores/uso terapéutico , Inflamación/inmunología , Linfocitos/inmunología , Melanoma/inmunología , Neoplasias Cutáneas/inmunología , Microambiente Tumoral , Adulto , Anciano , Anciano de 80 o más Años , Animales , Estudios de Casos y Controles , Línea Celular Tumoral , Técnicas de Cocultivo , Citocinas/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Voluntarios Sanos , Humanos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Inflamación/patología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Linfocitos/efectos de los fármacos , Melanoma/tratamiento farmacológico , Melanoma/metabolismo , Melanoma/patología , Ratones , Persona de Mediana Edad , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Adulto Joven
9.
Eur J Cancer ; 135: 173-182, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32590296

RESUMEN

BACKGROUND: Dendritic cells (DCs) are the most efficient antigen-presenting cells, hence initiating a potent and cancer-specific immune response. This ability (mainly using monocyte-derived DCs) has been exploited in vaccination strategies for decades with limited clinical efficacy. Another alternative would be the use of conventional DCs (cDCs) of which at least three subsets circulate in human blood: cDC1s (CD141bright), cDC2s (CD1c+) and plasmacytoid DCs. Despite their paucity, technical advances may allow for their selection and clinical use. However, many assumptions concerning the DC subset biology depend on observations from mouse models, hindering their translational potential. In this study, we characterise human DCs in patients with ovarian cancer (OvC) or prostate cancer (PrC). PATIENTS AND METHODS: Whole blood samples from patients with OvC or PrC and healthy donors (HDs) were evaluated by flow cytometry for the phenotypic and functional characterisation of DC subsets. RESULTS: In both patient groups, the frequency of total CD141+ DCs was lower than that in HDs, but the cDC1 subset was only reduced in patients with OvC. CD141+ DCs showed a reduced response to the TLR3 agonist poly (I:C) in both groups of patients. An inverse correlation between the frequency of cDC1s and CA125, the OvC tumour burden marker, was observed. Consistently, high expression of CLEC9A in OvC tissue (The Cancer Genome Atlas data set) indicated a better overall survival. CONCLUSIONS: cDC1s are reduced in patients with OvC, and CD141+ DCs are quantitatively and qualitatively impaired in patients with OvC or PrC. CD141+ DC activation may predict functional impairment. The loss of cDC1s may be a bad prognostic factor for patients with OvC.


Asunto(s)
Células Dendríticas/inmunología , Neoplasias Ováricas/inmunología , Neoplasias de la Próstata/inmunología , Anciano , Anciano de 80 o más Años , Antígenos de Superficie/sangre , Antígeno Ca-125/sangre , Estudios de Casos y Controles , Células Dendríticas/efectos de los fármacos , Femenino , Citometría de Flujo , Humanos , Inmunofenotipificación , Lectinas Tipo C/análisis , Masculino , Proteínas de la Membrana/sangre , Persona de Mediana Edad , Neoplasias Ováricas/sangre , Neoplasias Ováricas/mortalidad , Fenotipo , Poli I-C/farmacología , Pronóstico , Neoplasias de la Próstata/sangre , Neoplasias de la Próstata/mortalidad , Receptores Mitogénicos/análisis , Trombomodulina , Receptor Toll-Like 3/agonistas
10.
Front Immunol ; 10: 766, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31031762

RESUMEN

With the advent of combined immunotherapies, personalized dendritic cell (DC)-based vaccination could integrate the current standard of care for the treatment of a large variety of tumors. Due to their proficiency at antigen presentation, DC are key coordinators of the innate and adaptive immune system, and have critical roles in the induction of antitumor immunity. However, despite proven immunogenicity and favorable safety profiles, DC-based immunotherapies have not succeeded at inducing significant objective clinical responses. Emerging data suggest that the combination of DC-based vaccination with other cancer therapies may fully unleash the potential of DC-based cancer vaccines and improve patient survival. In this review, we discuss the recent efforts to develop innovative personalized DC-based vaccines and their use in combined therapies, with a particular focus on ovarian cancer and the promising results of mutanome-based personalized immunotherapies.


Asunto(s)
Vacunas contra el Cáncer , Células Dendríticas , Medicina de Precisión , Células Dendríticas/inmunología , Células Dendríticas/trasplante , Humanos , Inmunoterapia/métodos
11.
Front Immunol ; 10: 1845, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31456798

RESUMEN

T follicular helper (Tfh) cells have emerged as a critical limiting factor for controlling the magnitude of neonatal germinal center (GC) reactions and primary vaccine antibody responses. We compared the functional attributes of neonatal and adult Tfh cells at the transcriptomic level and demonstrated that the Tfh cell program is well-initiated in neonates although the Tfh gene-expression pattern (i.e., CXCR5, IL-21, BCL6, TBK1, STAT4, ASCL2, and c-MAF) is largely underrepresented as compared to adult Tfh cells. Importantly, we identified a TH2-bias of neonatal Tfh cells, with preferential differentiation toward short-lived pre-Tfh effector cells. Remarkably, adjuvantation with CpG-ODNs redirect neonatal pre-Tfh cells toward committed GC-Tfh cells, as illustrated by increased expression of Tfh signature genes and reduced expression of TH2-related genes.


Asunto(s)
Inmunidad Adaptativa , Centro Germinal/citología , Inmunidad Innata , Linfocitos T Colaboradores-Inductores/inmunología , Adyuvantes Inmunológicos , Envejecimiento/inmunología , Animales , Animales Recién Nacidos/inmunología , Centro Germinal/inmunología , Interleucina-13/metabolismo , Linfopoyesis/genética , Ratones Endogámicos C57BL , Células Th2/citología , Células Th2/inmunología , Transcriptoma
12.
Front Immunol ; 10: 1520, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31333656

RESUMEN

Pertussis is still observed in many countries despite of high vaccine coverage. Acellular pertussis (aP) vaccination is widely implemented in many countries as primary series in infants and as boosters in school-entry/adolescents/adults (including pregnant women in some). One novel strategy to improve the reactivation of aP-vaccine primed immunity could be to include genetically- detoxified pertussis toxin and novel adjuvants in aP vaccine boosters. Their preclinical evaluation is not straightforward, as it requires mimicking the human situation where T and B memory cells may persist longer than vaccine-induced circulating antibodies. Toward this objective, we developed a novel murine model including two consecutive adoptive transfers of the memory cells induced by priming and boosting, respectively. Using this model, we assessed the capacity of three novel aP vaccine candidates including genetically-detoxified pertussis toxin, pertactin, filamentous hemagglutinin, and fimbriae adsorbed to aluminum hydroxide, supplemented-or not-with Toll-Like-Receptor 4 or 9 agonists (TLR4A, TLR9A), to reactivate aP vaccine-induced immune memory and protection, reflected by bacterial clearance. In the conventional murine immunization model, TLR4A- and TLR9A-containing aP formulations induced similar aP-specific IgG antibody responses and protection against bacterial lung colonization as current aP vaccines, despite IL-5 down-modulation by both TLR4A and TLR9A and IL-17 up-modulation by TLR4A. In the absence of serum antibodies at time of boosting or exposure, TLR4A- and TLR9A-containing formulations both enhanced vaccine antibody recall compared to current aP formulations. Unexpectedly, however, protection was only increased by the TLR9A-containing vaccine, through both earlier bacterial control and accelerated clearance. This suggests that TLR9A-containing aP vaccines may better reactivate aP vaccine-primed pertussis memory and enhance protection than current or TLR4A-adjuvanted aP vaccines.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Bordetella pertussis/inmunología , Vacuna contra la Tos Ferina , Receptor Toll-Like 4 , Receptor Toll-Like 9 , Animales , Anticuerpos Antibacterianos/inmunología , Femenino , Ratones , Ratones Endogámicos BALB C , Vacuna contra la Tos Ferina/genética , Vacuna contra la Tos Ferina/inmunología , Vacuna contra la Tos Ferina/farmacología , Receptor Toll-Like 4/agonistas , Receptor Toll-Like 4/inmunología , Receptor Toll-Like 9/agonistas , Receptor Toll-Like 9/inmunología , Tos Ferina/inmunología , Tos Ferina/prevención & control
13.
Cell Rep ; 28(7): 1773-1784.e5, 2019 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-31412246

RESUMEN

Maternal antibodies (MatAbs) protect offspring from infections but limit their responses to vaccination. The mechanisms of this inhibition are still debated. Using murine early-life immunization models mimicking the condition prevailing in humans, we observed the induction of CD4-T, T follicular helper, and germinal center (GC) B cell responses even when early-life antibody responses were abrogated by MatAbs. GC B cells induced in the presence of MatAbs form GC structures and exhibit canonical GC changes in gene expression but fail to differentiate into plasma cells and/or memory B cells in a MatAb titer-dependent manner. Furthermore, GC B cells elicited in the presence or absence of MatAbs use different VH and Vk genes and show differences in genes associated with B cell differentiation and isotype switching. Thus, MatAbs do not prevent B cell activation but control the output of the GC reaction both quantitatively and qualitatively, shaping the antigen-specific B cell repertoire.


Asunto(s)
Anticuerpos Antivirales/inmunología , Anticuerpos/inmunología , Formación de Anticuerpos/inmunología , Linfocitos B/inmunología , Centro Germinal/inmunología , Orthomyxoviridae/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Animales Recién Nacidos , Anticuerpos/sangre , Anticuerpos Antivirales/sangre , Femenino , Inmunización , Masculino , Intercambio Materno-Fetal/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Embarazo , Vacunación
14.
J Immunother Cancer ; 7(1): 257, 2019 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-31601268

RESUMEN

BACKGROUND: Several mechanisms are present in the tumor microenvironment (TME) to impair cytotoxic T cell responses potentially able to control tumor growth. Among these, the accumulation of adenosine (Ado) contributes to tumor progression and represents a promising immunotherapeutic target. Ado has been shown to impair T cell effector function, but the role and mechanisms employed by Ado/Ado receptors (AdoRs) in modulating human peripheral and tumor-infiltrating lymphocyte (TIL) function are still puzzling. METHODS: CD8+ T cell cytokine production following stimulation was quantified by intracellular staining and flow cytometry. The cytotoxic capacity of tumor infiltrating lymphocytes (TILs) was quantified by the chromium release assay following co-culture with autologous or anti-CD3-loaded tumor cell lines. The CD8+ T cell metabolic fitness was evaluated by the seahorse assay and by the quantification of 2-NBDG uptake and CD71/CD98 upregulation upon stimulation. The expression of AdoRs was assessed by RNA flow cytometry, a recently developed technology that we validated by semiquantitative RT-PCR (qRT-PCR), while the impact on T cell function was evaluated by the use of selective antagonists and agonists. The influence of Ado/AdoR on the PKA and mTOR pathways was evaluated by phosphoflow staining of p-CREB and p-S6, respectively, and validated by western blot. RESULTS: Here, we demonstrate that Ado signaling through the A2A receptor (A2AR) in human peripheral CD8+ T cells and TILs is responsible for the higher sensitivity to Ado-mediated suppression of T central memory cells. We confirmed that Ado is able to impair peripheral and tumor-expanded T cell effector functions, and we show for the first time its impact on metabolic fitness. The Ado-mediated immunosuppressive effects are mediated by increased PKA activation that results in impairment of the mTORC1 pathway. CONCLUSIONS: Our findings unveil A2AR/PKA/mTORC1 as the main Ado signaling pathway impairing the immune competence of peripheral T cells and TILs. Thus, p-CREB and p-S6 may represent useful pharmacodynamic and efficacy biomarkers of immunotherapies targeting Ado. The effect of Ado on T cell metabolic fitness reinforces the importance of the adenosinergic pathway as a target for next-generation immunotherapy.


Asunto(s)
Adenosina/metabolismo , Linfocitos T CD8-positivos/inmunología , Neoplasias/inmunología , Transducción de Señal/inmunología , Microambiente Tumoral/inmunología , Adenosina/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Progresión de la Enfermedad , Femenino , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Persona de Mediana Edad , Neoplasias/metabolismo , Cultivo Primario de Células , Receptor de Adenosina A2A/metabolismo , Escape del Tumor
15.
Front Immunol ; 9: 381, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29541075

RESUMEN

Neonates and infants are more vulnerable to infections and show reduced responses to vaccination. Consequently, repeated immunizations are required to induce protection and early life vaccines against major pathogens such as influenza are yet unavailable. Formulating antigens with potent adjuvants, including immunostimulators and delivery systems, is a demonstrated approach to enhance vaccine efficacy. Yet, adjuvants effective in adults may not meet the specific requirements for activating the early life immune system. Here, we assessed the neonatal adjuvanticity of three novel adjuvants including TLR4 (glucopyranosyl lipid adjuvant-squalene emulsion), TLR9 (IC31®), and Mincle (CAF01) agonists, which all induce germinal centers (GCs) and potent antibody responses to influenza hemagglutinin (HA) in adult mice. In neonates, a single dose of HA formulated into each adjuvant induced T follicular helper (TFH) cells. However, only HA/CAF01 elicited significantly higher and sustained antibody responses, engaging neonatal B cells to differentiate into GCs already after a single dose. Although antibody titers remained lower than in adults, HA-specific responses induced by a single neonatal dose of HA/CAF01 were sufficient to confer protection against influenza viral challenge. Postulating that the neonatal adjuvanticity of CAF01 may result from the functionality of the C-type lectin receptor (CLR) Mincle in early life we asked whether other C-type lectin agonists would show a similar neonatal adjuvanticity. Replacing the Mincle agonist trehalose 6,6'-dibehenate by Curdlan, which binds to Dectin-1, enhanced antibody responses through the induction of similar levels of TFH, GCs and bone marrow high-affinity plasma cells. Thus, specific requirements of early life B cells may already be met after a single vaccine dose using CLR-activating agonists, identified here as promising B cell immunostimulators for early life vaccines when included into cationic liposomes.


Asunto(s)
Adyuvantes Inmunológicos , Linfocitos B/inmunología , Centro Germinal/inmunología , Glucolípidos/inmunología , Subtipo H1N1 del Virus de la Influenza A/fisiología , Gripe Humana/inmunología , Infecciones por Orthomyxoviridae/inmunología , beta-Glucanos/inmunología , Adyuvantes Inmunológicos/farmacología , Animales , Animales Recién Nacidos , Anticuerpos Antivirales/sangre , Femenino , Glucolípidos/farmacología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Humanos , Lectinas Tipo C/agonistas , Lectinas Tipo C/metabolismo , Liposomas , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Receptor Toll-Like 4/agonistas , Receptor Toll-Like 9/metabolismo , beta-Glucanos/farmacología
16.
Cancer Res ; 78(22): 6447-6461, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30185551

RESUMEN

In addition to improving insulin sensitivity in type 2 diabetes, the thiazolidinedione family of compounds and the pharmacologic activation of their best-characterized target PPARγ have been proposed as a therapeutic option for cancer treatment. In this study, we reveal a new mode of action for the thiazolidinedione rosiglitazone that can contribute to tumorigenesis. Rosiglitazone activated a tumorigenic paracrine communication program in a subset of human melanoma cells that involves the secretion of cytokines, chemokines, and angiogenic factors. This complex blend of paracrine signals activated nonmalignant fibroblasts, endothelial cells, and macrophages in a tumor-friendly way. In agreement with these data, rosiglitazone promoted human melanoma development in xenografts, and tumors exposed to rosiglitazone exhibited enhanced angiogenesis and inflammation. Together, these findings establish an important tumorigenic action of rosiglitazone in a subset of melanoma cells. Although studies conducted on cohorts of diabetic patients report overall benefits of thiazolidinediones in cancer prevention, our data suggest that exposure of established tumors to rosiglitazone may be deleterious.Significance: These findings uncover a novel mechanism by which the thiazolidinedione compound rosiglitazone contributes to tumorigenesis, thus highlighting a potential risk associated with its use in patients with established tumors. Cancer Res; 78(22); 6447-61. ©2018 AACR.


Asunto(s)
Melanoma/metabolismo , PPAR gamma/agonistas , Rosiglitazona/farmacología , Neoplasias Cutáneas/metabolismo , Células del Estroma/metabolismo , Inductores de la Angiogénesis/metabolismo , Animales , Carcinogénesis , Línea Celular Tumoral , Fibroblastos/metabolismo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Inflamación , Leucocitos Mononucleares/citología , Macrófagos/efectos de los fármacos , Melanoma/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Monocitos/metabolismo , Metástasis de la Neoplasia , Trasplante de Neoplasias , Análisis de Secuencia por Matrices de Oligonucleótidos , PPAR gamma/metabolismo , Comunicación Paracrina , Neoplasias Cutáneas/patología , Linfocitos T/citología
17.
J Exp Med ; 213(2): 177-87, 2016 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-26809444

RESUMEN

Evidence has recently emerged that butyrophilins, which are members of the extended B7 family of co-stimulatory molecules, have diverse functions in the immune system. We found that the human and mouse genes encoding butyrophilin-2A2 (BTN2A2) are regulated by the class II trans-activator and regulatory factor X, two transcription factors dedicated to major histocompatibility complex class II expression, suggesting a role in T cell immunity. To address this, we generated Btn2a2-deficient mice. Btn2a2(-/-) mice exhibited enhanced effector CD4(+) and CD8(+) T cell responses, impaired CD4(+) regulatory T cell induction, potentiated antitumor responses, and exacerbated experimental autoimmune encephalomyelitis. Altered immune responses were attributed to Btn2a2 deficiency in antigen-presenting cells rather than T cells or nonhematopoietic cells. These results provide the first genetic evidence that BTN2A2 is a co-inhibitory molecule that modulates T cell-mediated immunity.


Asunto(s)
Genes MHC Clase II , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Animales , Células Presentadoras de Antígenos/inmunología , Butirofilinas , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Línea Celular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Regulación de la Expresión Génica , Humanos , Inmunidad Celular , Glicoproteínas de Membrana/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Nucleares/genética , Proteínas Nucleares/inmunología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Transcripción del Factor Regulador X , Transactivadores/genética , Transactivadores/inmunología , Factores de Transcripción/genética , Factores de Transcripción/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA