Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Curr Top Microbiol Immunol ; 441: 21-59, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37695424

RESUMEN

Males and females differ in their susceptibility to develop autoimmunity and allergy but also in their capacity to cope with infections and cancers. Cellular targets and molecular pathways underlying sexual dimorphism in immunity have started to emerge and appeared multifactorial. It became increasingly clear that sex-linked biological factors have important impact on the development, tissue maintenance and effector function acquisition of distinct immune cell populations, thereby regulating multiple layers of innate or adaptive immunity through distinct mechanisms. This review discusses the recent development in our understanding of the cell-intrinsic actions of biological factors linked to sex, sex hormones and sex chromosome complement, on immune cells, which may account for the sex differences in susceptibility to autoimmune diseases and allergies, and the sex-biased responses in natural immunity and cancer.


Asunto(s)
Enfermedades Autoinmunes , Hipersensibilidad , Femenino , Masculino , Humanos , Cromosoma X , Hormonas Esteroides Gonadales/genética , Enfermedades Autoinmunes/genética , Inmunidad Adaptativa
2.
J Allergy Clin Immunol ; 149(1): 237-251.e12, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-33964300

RESUMEN

BACKGROUND: Allergic asthma is more severe and frequent in women than in men. In male mice, androgens negatively control group 2 innate lymphoid cell (ILC2) development and function by yet unknown mechanisms. OBJECTIVES: We sought to investigate the impact of androgen on ILC2 homeostasis and IL-33-mediated inflammation in female lungs. We evaluated the role of androgen receptor (AR) signaling and the contribution of the putative inhibitory receptor killer cell lectin-like receptor G1 (KLRG1). METHODS: Subcutaneous pellets mimicking physiological levels of androgen were used to treat female mice together with mice expressing a reporter enzyme under the control of androgen response elements and mixed bone marrow chimeras to assess the cell-intrinsic role of AR activation within ILC2s. We generated KLRG1-deficient mice. RESULTS: We established that lung ILC2s express a functionally active AR that can be in vivo targeted with exogenous androgens to negatively control ILC2 homeostasis, proliferation, and function. Androgen signaling upregulated KLRG1 on ILC2s, which inhibited their proliferation on E-cadherin interaction. Despite evidence that KLRG1 impaired the competitive fitness of lung ILC2s during inflammation, KLRG1 deficiency neither alters in vivo ILC2 numbers and functions, nor did it lead to hyperactive ILC2s in either sexes. CONCLUSIONS: AR agonists can be used in vivo to inhibit ILC2 homeostatic numbers and ILC2-dependent lung inflammation through cell-intrinsic AR activation. Although androgen signals in ILC2s to upregulate KLRG1, we demonstrate that KLRG1 is dispensable for androgen-mediated inhibition of pulmonary ILC2s.


Asunto(s)
Andrógenos/farmacología , Lectinas Tipo C/inmunología , Linfocitos/inmunología , Neumonía/inmunología , Receptores Inmunológicos/inmunología , Testosterona/farmacología , Animales , Femenino , Interleucina-33/inmunología , Pulmón/inmunología , Pulmón/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Neumonía/patología , Caracteres Sexuales , Transducción de Señal
3.
Eur J Immunol ; 49(11): 2030-2043, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31318439

RESUMEN

Plasmacytoid dendritic cells (pDCs) are found in the CNS during neuroinflammation and have been reported to exert regulatory functions in multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE). However, the mechanisms of entry of pDCs into the CNS as well as their phenotype and innate functional properties, once recruited into the CNS, have not been thoroughly examined. Herein, we show that pDCs rapidly accumulate into the brain and spinal cord during the acute phase of EAE, and maintain the expression of numerous phenotypic markers typical of peripheral pDCs. Functionally, CNS-pDCs constitutively expressed IRF7 and were able to rapidly produce type I IFNs and IL-12p40 upon ex vivo TLR-9 stimulation. Using adoptive transfer experiments, we provide evidence that CNS-pDC are recruited from the blood and accumulate into the CNS during the acute phase of EAE. Accumulation of pDCs into the CNS was strongly inhibited in the absence of CD29, but not CD18, suggesting a major role for ß1 but not ß2 integrins. Indeed, blocking the CD49d α4-integrins during acute EAE drastically diminished CNS-pDC numbers. Together, our results demonstrate that circulating pDCs are actively recruited into the CNS during acute EAE through a mechanism largely dependent on CD49d/CD29-integrins.


Asunto(s)
Encéfalo/inmunología , Células Dendríticas/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Integrina alfa4/inmunología , Integrina beta1/inmunología , Médula Espinal/inmunología , Traslado Adoptivo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/patología , Movimiento Celular/inmunología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/patología , Células Dendríticas/trasplante , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/patología , Femenino , Regulación de la Expresión Génica , Inmunidad Innata , Integrina alfa4/genética , Integrina beta1/genética , Factor 7 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/inmunología , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Subunidad p40 de la Interleucina-12/genética , Subunidad p40 de la Interleucina-12/inmunología , Ratones , Ratones Endogámicos C57BL , Glicoproteína Mielina-Oligodendrócito/administración & dosificación , Fragmentos de Péptidos/administración & dosificación , Toxina del Pertussis/administración & dosificación , Transducción de Señal , Médula Espinal/efectos de los fármacos , Médula Espinal/patología , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/inmunología
4.
J Immunol ; 201(11): 3218-3228, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30355786

RESUMEN

17ß-Estradiol (E2) suppresses the development of experimental autoimmune encephalomyelitis (EAE) through estrogen receptor (ER) α, yet the cellular targets remain elusive. We have used an adoptive transfer model of myelin oligodendrocyte glycoprotein-specific CD4+ T cells from 2D2 TCR transgenic mice. We show that in the recipient mice, ERα expression in bystander CD4+ T cells, rather than in cognate 2D2 T cells, is required for the inhibition of Th17 cell differentiation by E2. Coadministration of estrogen-primed WT, but not ERα-deficient CD4+ T cells, with naive 2D2 T cells lacking ERα inhibited the development of Th17 cell-mediated EAE. Suppression of Th17 cells and protection from EAE were maintained when ERα was deleted in Foxp3+ regulatory T cells. We showed that in vivo PD-L1 blockade alleviated the anti-inflammatory action of E2 and that PD-1 expression on cognate but not bystander T cells was required for the E2-dependent inhibition of Th17 differentiation. In cotransfer experiments, we found that only WT but not PD-1KO 2D2 T cells were amenable to E2-dependent inhibition of Th17 differentiation. These results support the conclusion that the restriction of Th17 cell development by E2-primed bystander CD4+ T cells requires cell-intrinsic PD-1 signaling within cognate T cells rather than induction of regulatory 2D2 T cells through PD-1 engagement. Altogether, our results indicate that pregnancy-level concentrations of estrogen signal in conventional Foxp3neg CD4+ T cells to limit the differentiation of cognate Th17 cells through a trans-acting mechanism of suppression that requires a functional PD-1/PD-L1 regulatory axis.


Asunto(s)
Efecto Espectador , Sistema Nervioso Central/inmunología , Estrógenos/metabolismo , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Traslado Adoptivo , Animales , Anticuerpos Monoclonales/administración & dosificación , Autoinmunidad , Antígeno B7-H1/inmunología , Antígenos CD4/metabolismo , Diferenciación Celular , Modelos Animales de Enfermedad , Receptor alfa de Estrógeno/genética , Femenino , Factores de Transcripción Forkhead/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Glicoproteína Mielina-Oligodendrócito/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Transducción de Señal
5.
Eur J Nucl Med Mol Imaging ; 46(7): 1506-1517, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30715571

RESUMEN

PURPOSE: This study sought to provide preliminary results on the biodistribution and dosimetry following intra-arterial liver injection of 188Re-SSS Lipiodol on hepatocellular carcinoma patients included in the Phase I Lip-Re 1 study. METHODS: Results of the first six patients included are reported. Analysis of the 188Re-SSS Lipiodol biodistribution was based on planar scintigraphic and tomoscintigraphic (SPECT) studies performed at 1, 6, 24, 48, and 72 h post-administration. Quantification in blood, urine, and stool samples was performed. Determination of the tumour to non-tumour uptake ratio (T/NT) was calculated. Absorbed doses to target organs and tumours were evaluated using the MIRD formalism. RESULTS: The mean injected activity of 188Re-SSS Lipiodol was 1645 ± 361 MBq. Uptakes were seen in the liver (tumour and healthy liver) and the lungs only. All these uptakes were stable over time. A mean 1.4 ± 0.7% of 188Re-SSS Lipiodol administered was detected in serum samples at 6 h, declining rapidly thereafter. On average, 1.5 ± 1.6% of administered activity was eliminated in urine and feces over 72 h. Overall, 90.7 ± 1.6% of detected activity on SPECT studies was found in the liver (74.9 ± 1.8% in tumours and 19.1 ± 1.7% in the healthy liver) and 9.3 ± 1.6% in the lungs (5.7 ± 1.1% in right and 3.7 ± 0.5% in left lungs). Mean doses absorbed were 7.9 ± 3.7Gy to the whole liver, 42.7 ± 34.0Gy to the tumours, 10.2 ± 3.7Gy to the healthy liver, and 1.5 ± 1.2Gy to the lungs. Four patients had stable disease on CT scans at 2 months. The first patient with rapidly progressive disease died at 1 month, most probably of massive tumour progression. Due to this early death and using a conservative approach, the trial independent evaluation committee decided to consider this event as a treatment-related toxicity. CONCLUSION: 188Re-SSS Lipiodol has a favorable biodistribution profile concerning radioembolization, with the highest in-vivo stability among all radiolabeled Lipiodol compounds reported to date. These preliminary results must be further confirmed while completing this Phase I Lip Re1 study.


Asunto(s)
Carcinoma Hepatocelular/diagnóstico por imagen , Carcinoma Hepatocelular/terapia , Embolización Terapéutica/métodos , Neoplasias Hepáticas/diagnóstico por imagen , Neoplasias Hepáticas/terapia , Hígado/diagnóstico por imagen , Anciano , Progresión de la Enfermedad , Combinación de Medicamentos , Femenino , Humanos , Inyecciones Intraarteriales , Aceite Yodado , Pulmón/diagnóstico por imagen , Masculino , Persona de Mediana Edad , Compuestos Organometálicos , Estudios Prospectivos , Radiometría , Radiofármacos/uso terapéutico , Distribución Tisular , Tomografía Computarizada de Emisión de Fotón Único , Tomografía Computarizada por Rayos X
6.
Immunity ; 32(4): 557-67, 2010 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-20399121

RESUMEN

Dendritic cells (DCs) play a pivotal role in controlling the balance between tolerance and immunity in the intestine. Gut conditioned CD103(+) DCs promote regulatory T (Treg) cell responses; however, little is known about DCs that drive inflammation in the intestine. Here, we show that monocyte-derived inflammatory DCs that express E-cadherin, the receptor for CD103, promote intestinal inflammation. E-cadherin(+) DCs accumulated in the inflamed mesenteric lymph nodes and colon, had high expression of toll-like receptors, and produced colitogenic cytokines, such as IL-6 and IL-23, after activation. Importantly, adoptive transfer of E-cadherin(+) DCs into T cell-restored immunodeficient hosts increased Th17 cell responses in the intestine and led to exacerbation of colitis. These results identify a monocyte-derived inflammatory DC subset that is associated with the pathogenesis of intestinal inflammation, providing a therapeutic target for the treatment of inflammatory bowel disease.


Asunto(s)
Cadherinas/inmunología , Colitis/inmunología , Células Dendríticas/inmunología , Linfocitos T/inmunología , Animales , Antígenos CD/inmunología , Antígeno CD11c/inmunología , Diferenciación Celular , Movimiento Celular , Colitis/genética , Colitis/patología , Células Dendríticas/citología , Regulación de la Expresión Génica , Inmunidad Innata , Inflamación/inmunología , Cadenas alfa de Integrinas/inmunología , Ratones , Ratones Endogámicos BALB C , Monocitos/citología , Monocitos/inmunología
7.
Eur J Nucl Med Mol Imaging ; 45(10): 1731-1741, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29560519

RESUMEN

PURPOSE: Selective internal radiation therapy (SIRT) appears to be an interesting treatment possibility for locally-advanced intrahepatic cholangiocarcinoma (ICC), yet the appropriate dosimetry has never been evaluated in this context. METHODS: We retrospectively studied data from 40 patients treated at our institution with 90Y-loaded glass microsphere SIRT combined with chemotherapy for inoperable ICC as first-line treatment. Macroaggregated albumin (MAA)-based single-photon emission computed tomography (SPECT)/computed tomography (CT) quantitative analysis was used to calculate the tumor dose (TD), healthy-injected liver dose (HILD), and injected liver dose (ILD). Response was evaluated at 3 months using the European Association for the Study of the Liver criteria. Factors associated with response and toxicity were analyzed using univariate analysis. RESULTS: We assessed a total of 35 patients (five excluded) receiving 55 injections. Mean TD was 322 ± 165Gy and mean HILD was 74 ± 24Gy for a mean ILD of 128 ± 28Gy. All but two lesions responded, with a minimal TD for responding lesions of 158Gy. Six Grade 3-4 permanent liver toxicities were observed. Mean HILD was not associated with liver toxicity (73.2 ± 25.8Gy for patients with liver toxicity and 77.8 ± 16.9Gy for patients without, ns). Only underlying Child-Pugh status (p = 0.0014) and underlying cirrhosis (p = 0.0021) were associated with liver toxicity. Median progression-free survival was 12.7 months and median overall survival (OS) was 28.6 months. Median OS was 52.7 months for patients with Child-Pugh A5 status. CONCLUSIONS: When combined with chemotherapy, SIRT is highly effective, with a TD > 158Gy. Tolerance was good except for the few patients with cirrhosis or Child-Pugh status ≥A6, who exhibited some liver toxicity. Prospective studies are warranted to confirm.


Asunto(s)
Albúminas/uso terapéutico , Neoplasias de los Conductos Biliares/tratamiento farmacológico , Neoplasias de los Conductos Biliares/radioterapia , Colangiocarcinoma/tratamiento farmacológico , Colangiocarcinoma/radioterapia , Vidrio/química , Microesferas , Albúminas/efectos adversos , Neoplasias de los Conductos Biliares/patología , Colangiocarcinoma/patología , Terapia Combinada , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Radiometría , Estudios Retrospectivos , Análisis de Supervivencia
8.
Eur J Nucl Med Mol Imaging ; 45(3): 392-401, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29177870

RESUMEN

PURPOSE: This study aimed at identifying prior therapy dosimetric parameters using 99mTc-labeled macro-aggregates of albumin (MAA) that are associated with contralateral hepatic hypertrophy occurring after unilobar radioembolization of hepatocellular carcinoma (HCC) performed with 90Y-loaded glass microspheres. METHODS: The dosimetry data of 73 HCC patients were collected prior to the treatment with 90Y-loaded microspheres for unilateral disease. The injected liver dose (ILD), the tumor dose (TD) and healthy injected liver dose (HILD) were calculated based on MAA quantification. Following treatment, the maximal hypertrophy (MHT) of an untreated lobe was calculated. RESULTS: Mean MHT was 35.4 ± 40.4%. When using continuous variables, the MHT was not correlated with any tested variable, i.e., injected activity, ILD, HILD or TD except with a percentage of future remnant liver (FRL) following the 90Y-microspheres injection  (r = -0.56). MHT ≥ 10% was significantly more frequent for patients with HILD ≥ 88 Gy, (52% of the cases), i.e., in 92.2% versus 65.7% for HILD < 88 Gy (p = 0.032). MHT ≥ 10% was also significantly more frequent for patients with a TD ≥ 205 Gy and a tumor volume (VT) ≥ 100 cm3 in patients with initial FRL < 50%. MHT ≥10% was seen in 83.9% for patients with either an HILD ≥ 88 Gy or a TD ≥ 205 Gy for tumors larger than 100cm3 (85% of the cases), versus only 54.5% (p = 0.0265) for patients with none of those parameters. MHT ≥10% was also associated with FRL and the Child-Pugh score. Using multivariate analysis, the Child-Pugh score (p < 0.0001), FRL (p = 0.0023) and HILD (p = 0.0029) were still significantly associated with MHT ≥10%. CONCLUSION: This study demonstrates for the first time that HILD is significantly associated with liver hypertrophy. There is also an impact of high tumor doses in large lesions in one subgroup of patients. Larger prospective studies evaluating the MAA dosimetric parameters have to be conducted to confirm these promising results.


Asunto(s)
Carcinoma Hepatocelular/radioterapia , Embolización Terapéutica/efectos adversos , Neoplasias Hepáticas/radioterapia , Hígado/patología , Hígado/efectos de la radiación , Radioisótopos de Itrio/efectos adversos , Radioisótopos de Itrio/uso terapéutico , Anciano , Femenino , Humanos , Hipertrofia/etiología , Masculino , Radiometría , Estudios Retrospectivos , Agregado de Albúmina Marcado con Tecnecio Tc 99m/efectos adversos , Agregado de Albúmina Marcado con Tecnecio Tc 99m/uso terapéutico
9.
Liver Int ; 37(1): 101-110, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27514012

RESUMEN

BACKGROUND & AIMS: Efficacy of radioembolization is derived from radioinduced damage, whereas tumour dosimetry is not considered as yet in prospective clinical trials. OBJECTIVES: This study evaluates the impact of tumour dose (TD), based on 99m Tc macroaggregated albumin (MAA) quantification, on response and overall survival (OS). MATERIALS AND METHODS: We consecutively included 85 patients with hepatocellular carcinoma treated with 90 Y-loaded glass microspheres. TD was calculated using a quantitative analysis of the MAA SPECT/CT. Responses were assessed after 3 months using the European Association for the Study of the Liver criteria. OS was assessed using Kaplan-Meier tests. RESULTS: Response rate was 80.3% on lesion-based analysis (n=132), and 77.5% on patient-based analysis. The response rate was only 9.1% for patients with TD <205 Gy against 89.7% for those with TD ≥205 Gy (P<10-7 ). Non-portal vein thrombosis (PVT) patients exhibited a median OS of 11.75 m (95% CI: 3-30.7 m) for TD <205 Gy, and 25 m (95% CI: 15-34.7 m) for TD ≥205 Gy (P=.0391). PVT patients exhibited a 4.35 m median OS (95% CI: 2-8 m) for TD<205 Gy, and 15.7 m (95% CI: 9.5-25.5 m) for TD ≥205 Gy, (P=.0004), with HR of 6.99. PVT patients exhibited a median OS of 3.6 m (95% CI: 2-8 m) when PVT MAA targeting was poor or with TD <205 Gy (poor candidate), vs 17.5 m (95% CI: 11-26.5 m) for the others identified as good candidates (P<.0001), with HR of 12.85. CONCLUSION: This study confirms the highly predictive value of MAA-based TD evaluation for response and OS. TD evaluation and PVT MAA targeting should be further evaluated in ongoing trials, whereas personalized dosimetry should be implemented in new trial designs.


Asunto(s)
Carcinoma Hepatocelular/terapia , Embolización Terapéutica , Neoplasias Hepáticas/terapia , Radiofármacos/uso terapéutico , Trombosis de la Vena/terapia , Anciano , Femenino , Francia , Humanos , Estimación de Kaplan-Meier , Modelos Logísticos , Masculino , Microesferas , Persona de Mediana Edad , Análisis Multivariante , Vena Porta/patología , Modelos de Riesgos Proporcionales , Estudios Prospectivos , Agregado de Albúmina Marcado con Tecnecio Tc 99m/uso terapéutico , Tomografía Computarizada por Rayos X , Trombosis de la Vena/patología , Radioisótopos de Itrio/uso terapéutico
10.
J Immunol ; 195(10): 4742-52, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26453746

RESUMEN

CD4(+) T cells polarize into effector Th subsets characterized by signature transcription factors and cytokines. Although T-bet drives Th1 responses and represses the alternative Th2, Th17, and Foxp3(+) regulatory T cell fates, the role of the T-bet-related transcription factor eomesodermin (Eomes) in CD4(+) T cells is less well understood. In this study, we analyze the expression and effects of Eomes in mouse CD4(+) T lymphocytes. We find that Eomes is readily expressed in activated CD4(+) Th1 T cells in vivo. Eomes(+) CD4(+) T cells accumulated in old mice, under lymphopenic conditions in a T cell transfer model of colitis, and upon oral Ag administration. However, despite its expression, genetic deletion of Eomes in CD4(+) T cells did not impact on IFN-γ production nor increase Th2 or Th17 responses. In contrast, Eomes deficiency favored the accumulation of Foxp3(+) cells in old mice, after in vivo differentiation of Eomes-deficient naive CD4(+) T cells, and in response to oral Ag in a cell-intrinsic way. Enforced Eomes expression during in vitro regulatory T cell induction also reduced Foxp3 transcription. Likewise, bystander Eomes-deficient CD4(+) T cells were more efficient at protecting from experimental autoimmune encephalitis compared with wild-type CD4(+) T cells. This enhanced capacity of Eomes-deficient CD4(+) T cells to inhibit EAE in trans was associated with an enhanced frequency of Foxp3(+) cells. Our data identify a novel role for Eomes in CD4(+) T cells and indicate that Eomes expression may act by limiting Foxp3 induction, which may contribute to the association of EOMES to susceptibility to multiple sclerosis.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Factores de Transcripción Forkhead/biosíntesis , Proteínas de Dominio T Box/fisiología , Envejecimiento/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Diferenciación Celular/inmunología , Modelos Animales de Enfermedad , Interferón gamma/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Dominio T Box/genética , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Células TH1/inmunología , Células TH1/metabolismo , Células Th17/inmunología , Células Th17/metabolismo , Células Th2/inmunología , Células Th2/metabolismo
11.
J Immunol ; 195(11): 5327-36, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26519527

RESUMEN

Increased IFN-α production contributes to the pathogenesis of infectious and autoimmune diseases. Plasmacytoid dendritic cells (pDCs) from females produce more IFN-α upon TLR7 stimulation than pDCs from males, yet the mechanisms underlying this difference remain unclear. In this article, we show that basal levels of IFN regulatory factor (IRF) 5 in pDCs were significantly higher in females compared with males and positively correlated with the percentage of IFN-α-secreting pDCs. Delivery of recombinant IRF5 protein into human primary pDCs increased TLR7-mediated IFN-α secretion. In mice, genetic ablation of the estrogen receptor 1 (Esr1) gene in the hematopoietic compartment or DC lineage reduced Irf5 mRNA expression in pDCs and IFN-α production. IRF5 mRNA levels furthermore correlated with ESR1 mRNA levels in human pDCs, consistent with IRF5 regulation at the transcriptional level by ESR1. Taken together, these data demonstrate a critical mechanism by which sex differences in basal pDC IRF5 expression lead to higher IFN-α production upon TLR7 stimulation in females and provide novel targets for the modulation of immune responses and inflammation.


Asunto(s)
Células Dendríticas/inmunología , Factores Reguladores del Interferón/metabolismo , Interferón-alfa/biosíntesis , Caracteres Sexuales , Receptor Toll-Like 7/metabolismo , Animales , Células Cultivadas , Receptor alfa de Estrógeno/genética , Femenino , Regulación de la Expresión Génica , Humanos , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/farmacología , Interferón-alfa/metabolismo , Masculino , Ratones , Ratones Transgénicos , ARN Mensajero/biosíntesis , Proteínas Recombinantes/farmacología , Transducción de Señal/genética
12.
Eur J Nucl Med Mol Imaging ; 43(3): 559-75, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26338177

RESUMEN

Radioembolization with (90)Y-loaded microspheres is increasingly used in the treatment of primary and secondary liver cancer. Technetium-99 m macroaggregated albumin (MAA) scintigraphy is used as a surrogate of microsphere distribution to assess lung or digestive shunting prior to therapy, based on tumoral targeting and dosimetry. To date, this has been the sole pre-therapeutic tool available for such evaluation. Several dosimetric approaches have been described using both glass and resin microspheres in hepatocellular carcinoma (HCC) and liver metastasis. Given that each product offers different specific activities and numbers of spheres injected, their radiobiological properties are believed to lightly differ. This paper summarizes and discusses the available studies focused on MAA-based dosimetry, particularly concentrating on potential confounding factors like clinical context, tumor size, cirrhosis, previous or concomitant therapy, and product used. In terms of the impact of tumoral dose in HCC, the results were concordant and a response relationship and tumoral threshold dose was clearly identified, especially in studies using glass microspheres. Tumoral dose has also been found to influence survival. The concept of treatment intensification has recently been introduced, yet despite several studies publishing interesting findings on the tumor dose-metastasis relationship, no consensus has been reached, and further clarification is thus required. Nor has the maximal tolerated dose to the liver been well documented, requiring more accurate evaluation. Lung dose was well described, despite recently identified factors influencing its evaluation, requiring further assessment. Conclusion: MAA SPECT/CT dosimetry is accurate in HCC and can now be used in order to achieve a fully customized approach, including treatment intensification. Yet further studies are warranted for the metastasis setting and evaluating the maximal tolerated liver dose.


Asunto(s)
Carcinoma Hepatocelular/diagnóstico por imagen , Carcinoma Hepatocelular/terapia , Neoplasias Hepáticas/diagnóstico por imagen , Neoplasias Hepáticas/terapia , Radioisótopos de Itrio/uso terapéutico , Angiografía , Embolización Terapéutica , Vidrio/química , Humanos , Hígado/diagnóstico por imagen , Pulmón/diagnóstico por imagen , Dosis Máxima Tolerada , Microesferas , Método de Montecarlo , Metástasis de la Neoplasia , Radiometría , Radiofármacos/uso terapéutico , Agregado de Albúmina Marcado con Tecnecio Tc 99m/química , Tomografía Computarizada de Emisión de Fotón Único , Tomografía Computarizada por Rayos X , Resultado del Tratamiento
13.
Eur J Nucl Med Mol Imaging ; 43(5): 824-831, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26686335

RESUMEN

PURPOSE: Radioembolization of liver cancer with (90)Y-loaded microspheres is increasingly used but data regarding hospital staff exposure are scarce. We evaluated the radiation exposure of medical staff while preparing and injecting (90)Y-loaded glass and resin microspheres especially in view of the increasing use of these products. METHODS: Exposure of the chest and finger of the radiopharmacist, nuclear medicine physician and interventional radiologist during preparation and injection of 78 glass microsphere preparations and 16 resin microsphere preparations was monitored. Electronic dosimeters were used to measure chest exposure and ring dosimeters were used to measure finger exposure. RESULTS: Chest exposure was very low for both products used (<10 µSv from preparation and injection). In our experience, finger exposure was significantly lower than the annual limit of 500 mSv for both products. With glass microspheres, the mean finger exposure was 13.7 ± 5.2 µSv/GBq for the radiopharmacist, and initially 17.9 ± 5.4 µSv/GBq for the nuclear medicine physician reducing to 13.97 ± 7.9 µSv/GBq with increasing experience. With resin microspheres, finger exposure was more significant: mean finger exposure for the radiopharmacist was 295.1 ± 271.9 µSv/GBq but with a reduction with increasing experience to 97.5 ± 35.2 µSv/GBq for the six most recent dose preparations. For administration of resin microspheres, the greatest mean finger exposure for the nuclear medicine physician (the most exposed operator) was 235.5 ± 156 µSv/GBq. CONCLUSION: Medical staff performing (90)Y-loaded microsphere radioembolization procedures are exposed to safe levels of radiation. Exposure is lower than that from treatments using (131)I-lipiodol. The lowest finger exposure is from glass microspheres. With resin microspheres finger exposure is acceptable but could be optimized in accordance with the ALARA principle, and especially in view of the increasing use of radioembolization.


Asunto(s)
Embolización Terapéutica/efectos adversos , Microesferas , Exposición Profesional/prevención & control , Exposición a la Radiación/prevención & control , Radiofármacos/efectos adversos , Radioterapia/efectos adversos , Radioisótopos de Itrio/efectos adversos , Adulto , Embolización Terapéutica/métodos , Dedos/efectos de la radiación , Humanos , Cuerpo Médico de Hospitales , Exposición Profesional/normas , Exposición a la Radiación/normas , Radiofármacos/administración & dosificación , Radiofármacos/uso terapéutico , Radioterapia/métodos , Torso/efectos de la radiación , Radioisótopos de Itrio/administración & dosificación , Radioisótopos de Itrio/uso terapéutico
14.
J Immunol ; 193(11): 5444-52, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25339659

RESUMEN

Human plasmacytoid dendritic cells (pDCs) play a major role in innate immunity through the production of type I IFNs after TLR engagement by pathogens. Sex-based differences in the innate function of human pDCs have been established, with pDCs from women exhibiting enhanced TLR7-mediated IFN-α production as compared with pDCs from males. In mice, we recently provided evidence for a role of estrogens as a positive regulator of pDC innate functions through cell-intrinsic estrogen receptor α signaling, but did not exclude a role for other X-linked factors, particularly in human pDCs. In this study, we investigated the respective contribution of X chromosome dosage and sex hormones using a humanized mouse model in which male or female NOD-SCID-ß2m(-/-) were transplanted with human progenitor cells purified from either male or female cord blood cells. We showed that, in response to TLR7 ligands, the frequency of IFN-α- and TNF-α-producing pDCs from either sex was greater in female than in male host mice, suggesting a positive role for estrogens. Indeed, blockade of estrogen receptor signaling during pDC development in vitro inhibited TLR7-mediated IFN-α production by human pDCs, which expressed both ESR1 and ESR2 genes. Interestingly, we also found that X chromosome dosage contributed to this sex bias as female pDCs have an enhanced TLR7-mediated IFN-α response as compared with male ones, irrespective of the sex of the recipient mice. Together, these results indicate that female sex hormones, estrogens, and X chromosome complement independently contribute to the enhanced TLR7-mediated IFN-α response of pDCs in women.


Asunto(s)
Células Dendríticas/fisiología , Receptor alfa de Estrógeno , Receptor beta de Estrógeno , Genes Ligados a X , Células Madre Hematopoyéticas/fisiología , Receptor Toll-Like 7/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Receptor alfa de Estrógeno/genética , Receptor beta de Estrógeno/genética , Femenino , Dosificación de Gen , Genes Ligados a X/genética , Trasplante de Células Madre Hematopoyéticas , Humanos , Inmunidad Innata , Interferón-alfa/metabolismo , Masculino , Ratones , Ratones SCID , Transducción de Señal/genética , Transducción de Señal/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
15.
J Immunol ; 190(11): 5459-70, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23626011

RESUMEN

17ß-Estradiol (E2) has been shown to regulate GM-CSF- or Flt3 ligand-driven dendritic cell (DC) development through estrogen receptor (ER) α signaling in myeloid progenitors. ERα regulates transcription of target genes through two distinct activation functions (AFs), AF-1 and AF-2, whose respective involvement varies in a cell type- or tissue-specific manner. In this study, we investigated the role of ERα AFs in the development and effector functions of inflammatory DCs, steady-state conventional DCs, and plasmacytoid DCs (pDC), using mouse lacking either AF-1 or AF-2. In agreement with previous works, we showed that E2 fostered the differentiation and effector functions of inflammatory DCs through ERα-dependent upregulation of IFN regulatory factor (IRF)-4 in GM-CSF-stimulated myeloid progenitors. Interestingly, whereas AF-1 was required for early IRF-4 upregulation in DC precursors, it was dispensable to enhance IRF-4 expression in differentiated DCs to a level compatible with the development of the more functional Ly6C(-) CD11b(+) DC subset. Presence of E2 had no effect on progenitors from either knock-in mice with 7-aa deletion in helix 12 of ERα, lacking AF-2, or ERα(-/-) mice. By contrast, in Flt3 ligand-driven DC differentiation, activation of AF-1 domain was required to promote the development of more functionally competent conventional DCs and pDCs. Moreover, lack of ERα AF-1 blunted the TLR7-mediated IFN-α response of female pDCs in vivo. Thus, our study demonstrates that ERα uses AF-1 differently in steady-state and inflammatory DC lineages to regulate their innate functions, suggesting that selective ER modulators could be used to target specific DC subsets.


Asunto(s)
Células Dendríticas/metabolismo , Estradiol/metabolismo , Receptor alfa de Estrógeno/metabolismo , Dominios y Motivos de Interacción de Proteínas , Animales , Antígenos CD11/metabolismo , Diferenciación Celular , Células Dendríticas/citología , Células Dendríticas/efectos de los fármacos , Receptor alfa de Estrógeno/química , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Masculino , Proteínas de la Membrana/farmacología , Ratones , Ratones Transgénicos , Fenotipo , Transducción de Señal , Receptores Toll-Like/metabolismo
16.
Blood ; 119(2): 454-64, 2012 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-22096248

RESUMEN

Plasmacytoid dendritic cells (pDCs) produce large amounts of type I interferons (IFN-α/ß) in response to viral or endogenous nucleic acids through activation of their endosomal Toll-like receptors (TLR-7 and TLR-9). Enhanced TLR-7-mediated IFN-α production by pDCs in women, compared with men, has been reported, but whether sex hormones, such as estrogens, are involved in this sex-based difference is unknown. Here we show, in humanized mice, that the TLR-7-mediated response of human pDCs is increased in female host mice relative to male. In a clinical trial, we establish that treatment of postmenopausal women with 17ß-estradiol markedly enhances TLR-7- and TLR-9-dependent production of IFN-α by pDCs stimulated by synthetic ligands or by nucleic acid-containing immune complexes. In mice, we found exogenous and endogenous estrogens to promote the TLR-mediated cytokine secretion by pDCs through hematopoietic expression of estrogen receptor (ER) α. Genetic ablation of ERα gene in the DC lineage abrogated the enhancing effect of 17ß-estradiol on their TLR-mediated production of IFN-α, showing that estrogens directly target pDCs in vivo. Our results uncover a previously unappreciated role for estrogens in regulating the innate functions of pDCs, which may account for sex-based differences in autoimmune and infectious diseases.


Asunto(s)
Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Estradiol/farmacología , Receptor alfa de Estrógeno/metabolismo , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 9/metabolismo , Adolescente , Adulto , Animales , Estudios de Casos y Controles , Citocinas/metabolismo , Células Dendríticas/metabolismo , Femenino , Humanos , Interferón Tipo I/metabolismo , Interferón-alfa/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Persona de Mediana Edad , Posmenopausia , Receptor Toll-Like 7/metabolismo , Adulto Joven
17.
Science ; 384(6692): eadk6200, 2024 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-38574174

RESUMEN

Males and females exhibit profound differences in immune responses and disease susceptibility. However, the factors responsible for sex differences in tissue immunity remain poorly understood. Here, we uncovered a dominant role for type 2 innate lymphoid cells (ILC2s) in shaping sexual immune dimorphism within the skin. Mechanistically, negative regulation of ILC2s by androgens leads to a reduction in dendritic cell accumulation and activation in males, along with reduced tissue immunity. Collectively, our results reveal a role for the androgen-ILC2-dendritic cell axis in controlling sexual immune dimorphism. Moreover, this work proposes that tissue immune set points are defined by the dual action of sex hormones and the microbiota, with sex hormones controlling the strength of local immunity and microbiota calibrating its tone.


Asunto(s)
Andrógenos , Células Dendríticas , Inmunidad Innata , Linfocitos , Caracteres Sexuales , Piel , Femenino , Masculino , Andrógenos/metabolismo , Células Dendríticas/inmunología , Hormonas Esteroides Gonadales/metabolismo , Linfocitos/inmunología , Piel/inmunología , Animales , Ratones , Ratones Endogámicos C57BL , Microbiota
18.
J Immunol ; 187(5): 2386-93, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21810607

RESUMEN

Estrogen treatment exerts a protective effect on experimental autoimmune encephalomyelitis (EAE) and is under clinical trial for multiple sclerosis therapy. Estrogens have been suspected to protect from CNS autoimmunity through their capacity to exert anti-inflammatory as well as neuroprotective effects. Despite the obvious impacts of estrogens on the pathophysiology of multiple sclerosis and EAE, the dominant cellular target that orchestrates the anti-inflammatory effect of 17ß-estradiol (E2) in EAE is still ill defined. Using conditional estrogen receptor (ER) α-deficient mice and bone marrow chimera experiments, we show that expression of ERα is critical in hematopoietic cells but not in endothelial ones to mediate the E2 inhibitory effect on Th1 and Th17 cell priming, resulting in EAE protection. Furthermore, using newly created cell type-specific ERα-deficient mice, we demonstrate that ERα is required in T lymphocytes, but neither in macrophages nor dendritic cells, for E2-mediated inhibition of Th1/Th17 cell differentiation and protection from EAE. Lastly, in absence of ERα in host nonhematopoietic tissues, we further show that ERα signaling in T cells is necessary and sufficient to mediate the inhibitory effect of E2 on EAE development. These data uncover T lymphocytes as a major and nonredundant cellular target responsible for the anti-inflammatory effects of E2 in Th17 cell-driven CNS autoimmunity.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Estradiol/farmacología , Receptor alfa de Estrógeno/metabolismo , Estrógenos/farmacología , Células TH1/efectos de los fármacos , Células Th17/efectos de los fármacos , Animales , Southern Blotting , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Separación Celular , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Receptor alfa de Estrógeno/inmunología , Femenino , Citometría de Flujo , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Células TH1/citología , Células TH1/inmunología , Células Th17/citología , Células Th17/inmunología
19.
Cancers (Basel) ; 15(8)2023 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-37190173

RESUMEN

BACKGROUND: Despite the wide development of 90Y-loaded microspheres, 188Re-labeled lipiodol is still being used for radioembolization of hepatocellular carcinoma (HCC). However, the use of this latter compound is limited by in vivo instability. This study sought to evaluate the safety, bio-distribution, and response to 188Re-SSS lipiodol, a new and more stable compound. METHOD: Lip-Re-01 was an activity-escalation Phase 1 study involving HCC patients progressing after sorafenib. The primary endpoint was safety based on Common Terminology Criteria for Adverse Events (AEs) of Grade ≥3 within 2 months. Secondary endpoints included bio-distribution assessed by scintigraphy quantification from 1 to 72 h, tumor to non-tumor uptake ratio (T/NT), as well as blood, urine and feces collection over 72 h, dosimetry, and response evaluation (mRECIST). RESULTS: Overall, 14 heavily pre-treated HCC patients were treated using a whole liver approach. The mean injected activity was 1.5 ± 0.4 GBq for activity Level 1 (n = 6), 3.6 ± 0.3 GBq for Level 2 (n = 6), and 5.0 ± 0.4 GBq for Level 3 (n = 2). Safety was acceptable with only 1/6 of Level 1 and 1/6 of Level 2 patients experiencing limiting toxicity (one liver failure; one lung disease). The study was prematurely discontinued unrelated to clinical outcomes. Uptake occurred in the tumor, liver, and lungs, and only sometimes in the bladder. The T/NT ratio was high with a mean of 24.9 ± 23.4. Cumulative urinary elimination and fecal eliminations at 72 h were very low, 4.8 ± 3.2% and 0.7 ± 0.8%, respectively. Partial response occurred in 21% of patients (0% in the first activity level; 37.5% in the others). CONCLUSION: The high in vivo stability of 188Re-SSS lipiodol was confirmed, resulting in encouraging responses for a Phase 1 study. As the 3.6 GBq activity proved to be safe, it will be used in a future Phase 2 study.

20.
EBioMedicine ; 80: 104047, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35561451

RESUMEN

BACKGROUND: Type I interferon (IFN-I) production by plasmacytoid dendritic cells (pDCs) occurs during viral infection, in response to Toll-like receptor 7 (TLR7) stimulation and is more vigorous in females than in males. Whether this sex bias persists in ageing people is currently unknown. In this study, we investigated the effect of sex and aging on IFN-α production induced by PRR agonist ligands. METHODS: In a large cohort of individuals from 19 to 97 years old, we measured the production of IFN-α and inflammatory cytokines in whole-blood upon stimulation with either R-848, ODN M362 CpG-C, or cGAMP, which activate the TLR7/8, TLR9 or STING pathways, respectively. We further characterized the cellular sources of IFN-α. FINDINGS: We observed a female predominance in IFN-α production by pDCs in response to TLR7 or TLR9 ligands. The higher TLR7-driven IFN-α production in females was robustly maintained across ages, including the elderly. The sex-bias in TLR9-driven interferon production was lost after age 60, which correlated with the decline in circulating pDCs. By contrast, STING-driven IFN-α production was similar in both sexes, preserved with aging, and correlated with circulating monocyte numbers. Indeed, monocytes were the primary cellular source of IFN-α in response to cGAMP. INTERPRETATION: We show that the sex bias in the TLR7-induced IFN-I production is strongly maintained through ages, and identify monocytes as the main source of IFN-I production via STING pathway. FUNDING: This work was supported by grants from Région Occitanie/Pyrénées-Méditerranée (#12052910, Inspire Program #1901175), University Paul Sabatier, and the European Regional Development Fund (MP0022856).


Asunto(s)
Interferón-alfa , Monocitos , Receptor Toll-Like 7 , Adulto , Anciano , Anciano de 80 o más Años , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Femenino , Humanos , Interferón-alfa/biosíntesis , Interferón-alfa/sangre , Interferón-alfa/inmunología , Ligandos , Masculino , Proteínas de la Membrana/sangre , Proteínas de la Membrana/inmunología , Persona de Mediana Edad , Monocitos/inmunología , Monocitos/metabolismo , Receptor Toll-Like 9/metabolismo , Adulto Joven
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda