Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Nat Commun ; 15(1): 1718, 2024 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-38409097

RESUMEN

Foxo family transcription factors are critically involved in multiple processes, such as metabolism, quiescence, cell survival and cell differentiation. Although continuous, high activity of Foxo transcription factors extends the life span of some species, the involvement of Foxo proteins in mammalian aging remains to be determined. Here, we show that Foxo1 is down-regulated with age in mouse T cells. This down-regulation of Foxo1 in T cells may contribute to the disruption of naive T-cell homeostasis with age, leading to an increase in the number of memory T cells. Foxo1 down-regulation is also associated with the up-regulation of co-inhibitory receptors by memory T cells and exhaustion in aged mice. Using adoptive transfer experiments, we show that the age-dependent down-regulation of Foxo1 in T cells is mediated by T-cell-extrinsic cues, including type 1 interferons. Taken together, our data suggest that type 1 interferon-induced Foxo1 down-regulation is likely to contribute significantly to T-cell dysfunction in aged mice.


Asunto(s)
Factores de Transcripción Forkhead , Agotamiento de Células T , Ratones , Animales , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Regulación hacia Abajo , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Diferenciación Celular , Proteínas/metabolismo , Interferones/metabolismo , Mamíferos/metabolismo
2.
Surg Endosc ; 38(2): 769-779, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38052888

RESUMEN

BACKGROUND: Three randomized controlled trials have reported improved functional recovery after Laparoscopic pancreatoduodenectomy (LPD), as compared to open pancreatoduodenectomy (OPD). Long-term results regarding quality of life (QoL) are lacking. The aim of this study was to compare long-term QoL of LPD versus OPD. METHODS AND PATIENTS: A monocentric retrospective cross-sectional study was performed among patients < 75 years old who underwent LPD or OPD for a benign or premalignant pathology in a high-volume center (2011-2021). An electronic three-part questionnaire was sent to eligible patients, including two diseases specific QoL questionnaires (the European Organization for Research and Treatment in Cancer Quality of Life Questionnaire for cancer (QLQ-C30) and a pancreatic cancer module (PAN26) and a body image questionnaire. Patient demographics and postoperative data were collected and compared between LPD and OPD. RESULTS: Among 948 patients who underwent PD (137 LPD, 811 OPD), 170 were eligible and 111 responded (58 LPD and 53 OPD). LPD versus OPD showed no difference in mean age (51 vs. 55 years, p = 0.199) and female gender (40% vs. 45%, p = 0.631), but LPD showed lower BMI (24 vs 26; p = 0.028) and higher preoperative pancreatitis (29% vs 13%; p = 0.041). The postoperative outcome showed similar Clavien-Dindo ≥ III morbidity (19% vs. 23%; p = 0.343) and length of stay (24 vs. 21 days, p = 0.963). After a similar median follow-up (3 vs. 3 years; p = 0.122), LPD vs OPD patients reported higher QoL (QLQ-C30: 49.6 vs 56.3; p = 0.07), better pancreas specific health status score (PAN20: 50.5 vs 55.5; p = 0.002), physical functioning (p = 0.002), and activities limitations (p = 0.02). Scar scores were better after LPD regarding esthetics (p = 0.001), satisfaction (p = 0.04), chronic pain at rest (p = 0.036), moving (p = 0.011) or in daily activities (p = 0.02). There was no difference in digestive symptoms (p = 0.995). CONCLUSION: This monocentric study found improved long-term QoL in patients undergoing LPD, as compared to OPD, for benign and premalignant diseases. These results could be considered when choosing the surgical approach in these patients.


Asunto(s)
Laparoscopía , Neoplasias Pancreáticas , Humanos , Femenino , Anciano , Pancreaticoduodenectomía/métodos , Calidad de Vida , Estudios Retrospectivos , Estudios Transversales , Tiempo de Internación , Neoplasias Pancreáticas/cirugía , Laparoscopía/métodos , Complicaciones Posoperatorias/epidemiología , Complicaciones Posoperatorias/etiología , Complicaciones Posoperatorias/cirugía
3.
EBioMedicine ; 91: 104578, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-37086650

RESUMEN

BACKGROUND: Estimation of glomerular function is necessary to diagnose kidney diseases. However, the study of glomeruli in the clinic is currently done indirectly through urine and blood tests. A recent imaging technique called Ultrasound Localization Microscopy (ULM) has appeared. It is based on the ability to record continuous movements of individual microbubbles in the bloodstream. Although ULM improved the resolution of vascular imaging up to tenfold, the imaging of the smallest vessels had yet to be reported. METHODS: We acquired ultrasound sequences from living humans and rats and then applied filters to divide the data set into slow-moving and fast-moving microbubbles. We performed a double tracking to highlight and characterize populations of microbubbles with singular behaviors. We decided to call this technique "sensing ULM" (sULM). We used post-mortem micro-CT for side-by-side confirmation in rats. FINDINGS: In this study, we report the observation of microbubbles flowing in the glomeruli in living humans and rats. We present a set of analysis tools to extract quantitative information from individual microbubbles, such as remanence time or normalized distance. INTERPRETATION: As glomeruli play a key role in kidney function, it would be possible that their observation yields a deeper understanding of the kidney. It could also be a tool to diagnose kidney diseases in patients. More generally, it will bring imaging capabilities closer to the functional units of organs, which is a key to understand most diseases, such as cancer, diabetes, or kidney failures. FUNDING: This study was funded by the European Research Council under the European Union Horizon H2020 program (ERC Consolidator grant agreement No 772786-ResolveStroke).


Asunto(s)
Enfermedades Renales , Microscopía , Humanos , Ratas , Animales , Microscopía/métodos , Ultrasonografía/métodos , Glomérulos Renales/diagnóstico por imagen , Riñón/diagnóstico por imagen , Medios de Contraste
4.
J Hepatol ; 77(2): 424-435, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35257829

RESUMEN

BACKGROUND & AIMS: One-third of hepatocellular carcinomas (HCCs) harbor mutations activating the ß-catenin pathway, predominantly via mutations in the CTNNB1 gene itself. Mouse models of Apc loss-of-function are widely used to mimic ß-catenin-dependent tumorigenesis. Given the low prevalence of APC mutations in human HCCs, we aimed to generate liver tumors through CTNNB1 exon 3 deletion (ßcatΔex3). We then compared ßcatΔex3 liver tumors with liver tumors generated via frameshift in exon 15 of Apc (Apcfs-ex15). METHODS: We used hepatocyte-specific and inducible mouse models generated through either a Cre-Lox or a CRISPR/Cas9 approach using adeno-associated virus vectors. Tumors generated by the Cre-Lox models were phenotypically analyzed using immunohistochemistry and were selected for transcriptomic analysis by RNA-sequencing (RNAseq). Mouse RNAseq data were compared to human RNAseq data (8 normal tissues, 48 HCCs, 9 hepatoblastomas) in an integrative analysis. Tumors generated via CRISPR were analyzed using DNA sequencing and immuno-histochemistry. RESULTS: Mice with CTNNB1 exon 3 deletion in hepatocytes developed liver tumors indistinguishable from Apcfs-ex15 liver tumors. Both Apcfs-ex15 and ßcatΔex3 mouse models induced growth of phenotypically distinct tumors (differentiated or undifferentiated). Integrative analysis of human and mouse tumors showed that differentiated mouse tumors cluster with well-differentiated human CTNNB1-mutated tumors. Conversely, undifferentiated mouse tumors cluster with human mesenchymal hepatoblastomas and harbor activated YAP signaling. CONCLUSION: Apcfs-ex15 and ßcatΔex3 mouse models both induce growth of tumors that are transcriptionally similar to either well-differentiated and ß-catenin-activated human HCCs or mesenchymal hepatoblastomas. LAY SUMMARY: New and easy-to-use transgenic mouse models of primary liver cancers have been generated, with mutations in the gene encoding beta-catenin, which are frequent in both adult and pediatric primary liver cancers. The mice develop both types of cancer, constituting a strong preclinical model.


Asunto(s)
Carcinoma Hepatocelular , Hepatoblastoma , Neoplasias Hepáticas , beta Catenina , Animales , Carcinoma Hepatocelular/patología , Hepatoblastoma/metabolismo , Hepatocitos/metabolismo , Humanos , Neoplasias Hepáticas/patología , Ratones , Ratones Transgénicos , Mutación , beta Catenina/genética
5.
Elife ; 102021 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-34106045

RESUMEN

Only a fraction of cancer patients benefits from immune checkpoint inhibitors. This may be partly due to the dense extracellular matrix (ECM) that forms a barrier for T cells. Comparing five preclinical mouse tumor models with heterogeneous tumor microenvironments, we aimed to relate the rate of tumor stiffening with the remodeling of ECM architecture and to determine how these features affect intratumoral T cell migration. An ECM-targeted strategy, based on the inhibition of lysyl oxidase, was used. In vivo stiffness measurements were found to be strongly correlated with tumor growth and ECM crosslinking but negatively correlated with T cell migration. Interfering with collagen stabilization reduces ECM content and tumor stiffness leading to improved T cell migration and increased efficacy of anti-PD-1 blockade. This study highlights the rationale of mechanical characterizations in solid tumors to understand resistance to immunotherapy and of combining treatment strategies targeting the ECM with anti-PD-1 therapy.


Asunto(s)
Fenómenos Fisiológicos Celulares/efectos de los fármacos , Colágeno/metabolismo , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Linfocitos T/metabolismo , Microambiente Tumoral/fisiología , Animales , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Matriz Extracelular/metabolismo , Femenino , Inhibidores de Puntos de Control Inmunológico/farmacología , Ratones , Ratones Endogámicos C57BL , Neoplasias Experimentales , Proteína-Lisina 6-Oxidasa/metabolismo
6.
Theranostics ; 11(8): 3830-3838, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33664864

RESUMEN

Anti-angiogenics drugs in clinical use for cancer treatment induce cardiotoxic side effects. The endothelin axis is involved in hypertension and cardiac remodelling, and addition of an endothelin receptor antagonist to the anti-angiogenic sunitinib was shown to reduce cardiotoxicity of sunitinib in mice. Here, we explored further the antidote effect of the endothelin receptor antagonist macitentan in sunitinib-treated animals on cardiac remodeling. Methods: Tumor-bearing mice treated per os daily by sunitinib or vehicle were imaged before and after 1, 3 and 6 weeks of treatment by positron emission tomography using [18F]fluorodeoxyglucose and by echocardiography. Non-tumor-bearing animals were randomly assigned to be treated per os daily by vehicle or sunitinib or macitentan or sunitinib+macitentan, and imaged by echocardiography after 5 weeks. Hearts were harvested for histology and molecular analysis at the end of in vivo exploration. Results: Sunitinib treatment increases left ventricular mass and ejection fraction and induces cardiac fibrosis. Sunitinib also induces an early increase in cardiac uptake of [18F]fluorodeoxyglucose, which is significantly correlated with increased left ventricular mass at the end of treatment. Co-administration of macitentan prevents sunitinib-induced hypertension, increase in ejection fraction and cardiac fibrosis, but fails to prevent increase of the left ventricular mass. Conclusion: Early metabolic changes predict sunitinib-induced cardiac remodeling. Endothelin blockade can prevent some but not all cardiotoxic side-effects of sunitinib, in particular left ventricle hypertrophy that appears to be induced by sunitinib through an endothelin-independent mechanism.


Asunto(s)
Cardiomegalia/inducido químicamente , Endotelinas/fisiología , Sunitinib/toxicidad , Animales , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Modelos Animales de Enfermedad , Antagonistas de los Receptores de Endotelina/administración & dosificación , Femenino , Fibrosis , Glucólisis/efectos de los fármacos , Hipertensión/inducido químicamente , Hipertensión/prevención & control , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Medicina de Precisión , Pirimidinas/administración & dosificación , Sulfonamidas/administración & dosificación , Remodelación Ventricular/efectos de los fármacos , Remodelación Ventricular/fisiología
7.
Sci Rep ; 11(1): 4219, 2021 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-33603134

RESUMEN

Women diagnosed with high-grade serous ovarian cancers (HGSOC) are still likely to exhibit a bad prognosis, particularly when suffering from HGSOC of the Mesenchymal molecular subtype (50% cases). These tumors show a desmoplastic reaction with accumulation of extracellular matrix proteins and high content of cancer-associated fibroblasts. Using patient-derived xenograft mouse models of Mesenchymal and Non-Mesenchymal HGSOC, we show here that HGSOC exhibit distinct stiffness depending on their molecular subtype. Indeed, tumor stiffness strongly correlates with tumor growth in Mesenchymal HGSOC, while Non-Mesenchymal tumors remain soft. Moreover, we observe that tumor stiffening is associated with high stromal content, collagen network remodeling, and MAPK/MEK pathway activation. Furthermore, tumor stiffness accompanies a glycolytic metabolic switch in the epithelial compartment, as expected based on Warburg's effect, but also in stromal cells. This effect is restricted to the central part of stiff Mesenchymal tumors. Indeed, stiff Mesenchymal tumors remain softer at the periphery than at the core, with stromal cells secreting high levels of collagens and showing an OXPHOS metabolism. Thus, our study suggests that tumor stiffness could be at the crossroad of three major processes, i.e. matrix remodeling, MEK activation and stromal metabolic switch that might explain at least in part Mesenchymal HGSOC aggressiveness.


Asunto(s)
Colágeno/metabolismo , Mesodermo/metabolismo , Mesodermo/patología , Miofibroblastos/metabolismo , Miofibroblastos/patología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Animales , Línea Celular Tumoral , Cistadenocarcinoma Seroso/metabolismo , Cistadenocarcinoma Seroso/patología , Femenino , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Células del Estroma/metabolismo , Células del Estroma/patología
8.
ACS Nano ; 14(5): 5738-5753, 2020 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-32338871

RESUMEN

Physical oncology recognizes tissue stiffness mediated by activation of cancer-associated fibroblasts (CAF) and extracellular matrix remodeling as an active modulator of tumorigenesis, treatment resistance, and clinical outcome. Cholangiocarcinoma (CCA) is a highly aggressive and chemoresistant desmoplastic cancer enriched in CAF. CCA's stroma mechanical properties are considered responsible for its chemoresistant character. To normalize tumor mechanics, we propose a physical strategy based on remotely light-activated nanohyperthermia to modulate the tumor microenvironment. In this study, we report the use of multifunctional iron oxide nanoflowers decorated with gold nanoparticles (GIONF) as efficient nanoheaters to achieve complete tumor regression following three sessions of mild hyperthermia. The preferential uptake of GIONF by CAF allowed targeting this cell population, which resulted in a significant early reduction of tumor stiffness followed by tumor regression. In conclusion, our study highlights a spatially and temporally controlled physical strategy, GIONF-mediated photothermal therapy to deplete CAF and normalize the tumor mechanics that may apply to desmoplastic cancer and CCA treatment.


Asunto(s)
Neoplasias de los Conductos Biliares , Fibroblastos Asociados al Cáncer , Colangiocarcinoma , Nanopartículas del Metal , Neoplasias de los Conductos Biliares/terapia , Conductos Biliares Intrahepáticos , Colangiocarcinoma/terapia , Fibroblastos , Oro , Humanos , Microambiente Tumoral
9.
J Exp Med ; 216(11): 2669-2687, 2019 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-31492810

RESUMEN

Neutrophils produce high levels of reactive oxygen species (ROS) by NADPH oxidase that are crucial for host defense but can lead to tissue injury when produced in excess. We previously described that proliferating cell nuclear antigen (PCNA), a nuclear scaffolding protein pivotal in DNA synthesis, controls neutrophil survival through its cytosolic association with procaspases. We herein showed that PCNA associated with p47phox, a key subunit of NADPH oxidase, and that this association regulated ROS production. Surface plasmon resonance and crystallography techniques demonstrated that the interdomain-connecting loop of PCNA interacted directly with the phox homology (PX) domain of the p47phox. PCNA inhibition by competing peptides or by T2AA, a small-molecule PCNA inhibitor, decreased NADPH oxidase activation in vitro. Furthermore, T2AA provided a therapeutic benefit in mice during trinitro-benzene-sulfonic acid (TNBS)-induced colitis by decreasing oxidative stress, accelerating mucosal repair, and promoting the resolution of inflammation. Our data suggest that targeting PCNA in inflammatory neutrophils holds promise as a multifaceted antiinflammatory strategy.


Asunto(s)
Citosol/metabolismo , NADPH Oxidasa 2/metabolismo , NADPH Oxidasas/metabolismo , Neutrófilos/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Animales , Línea Celular Tumoral , Células Cultivadas , Colitis/inducido químicamente , Colitis/prevención & control , Activación Enzimática/efectos de los fármacos , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , NADPH Oxidasas/genética , Unión Proteica , Especies Reactivas de Oxígeno/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Ácido Trinitrobencenosulfónico
10.
Clin Res Hepatol Gastroenterol ; 43(6): 669-681, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31031131

RESUMEN

Targeted and triggered release of liposomal drug using ultrasound (US) induced cavitation represents a promising treatment modality to increase the therapeutic-toxicity ratio of encapsulated chemotherapy. OBJECTIVES: To study the feasibility and efficacy of a combination of focused US and liposomal doxorubicin (US-L-DOX) release in orthotopic murine models of pancreatic cancer. MATERIAL AND METHODS: A confocal US setup was developed to generate US inertial cavitation delivery in a controlled and reproducible manner and designed for two distinct murine orthotopic pancreatic cancer models. Controlled cavitation at 1 MHz was applied within the tumors after L-DOX injection according to a preliminary pharmacokinetic study. RESULTS: In vitro studies confirmed that L-DOX was cytostatic. In vivo pharmacokinetic study showed L-DOX peak tumor accumulation at 48h. Feasibility of L-DOX injection and US delivery was demonstrated in both murine models. In a nude mouse model, at W9 after implantation (W5 after treatment), US-L-DOX group (median [IQR] 51.43 mm3 [35.1-871.95]) exhibited significantly lower tumor volumes than the sham group (216.28 [96.12-1202.92]), the US group (359.44 [131.48-1649.25]), and the L-DOX group (255.94 [84.09-943.72]), and a trend, although not statistically significant, to a lower volume than Gemcitabine group (90.48 [42.14-367.78]). CONCLUSION: This study demonstrates that inertial cavitation can be generated to increase the therapeutic effect of drug-carrying liposomes accumulated in the tumor. This approach is potentially an important step towards a therapeutic application of cavitation-induced drug delivery in pancreatic cancer.


Asunto(s)
Antibióticos Antineoplásicos/administración & dosificación , Doxorrubicina/análogos & derivados , Neoplasias Pancreáticas/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Doxorrubicina/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Estudios de Factibilidad , Femenino , Liposomas , Masculino , Ratones , Ratones Desnudos , Polietilenglicoles/administración & dosificación , Ratas , Ratas Endogámicas Lew , Ultrasonografía
11.
Methods Mol Biol ; 1943: 377-387, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30838630

RESUMEN

Ultrasound-mediated gene delivery is an interesting approach, which could help in increasing gene transfer in deep tissues. Moreover, it allows for performing experiments guided by the image to determine which elements are required. Microbubbles complexed with a eukaryotic expression cassette are excellent agents as they are responsive to ultrasounds and, upon oscillation, can destabilize membranes to enhance gene transfer. Here, we describe the preparation of positively charged microbubbles, plasmid free of antibiotic resistance marker, their combination and the conditions of ultrasound-mediated liver transfection post-systemic administration in mice. This association allowed us to obtain a superior liver gene expression at least over 8 months after a single injection.


Asunto(s)
Microburbujas , Transfección/métodos , Ondas Ultrasónicas , Animales , Permeabilidad de la Membrana Celular/efectos de la radiación , Terapia Genética/métodos , Células HeLa , Humanos , Hígado/citología , Hígado/metabolismo , Hígado/efectos de la radiación , Ratones , Ratones Endogámicos BALB C , Ácidos Nucleicos/genética
12.
Liver Int ; 39(4): 727-739, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30721564

RESUMEN

BACKGROUND & AIMS: Loss of hepatocyte nuclear factor-4α (HNF4α), a critical factor driving liver development and differentiation, is frequently associated with hepatocellular carcinoma (HCC). Our recent data revealed that HNF4α level was decreased in mouse and human HCCs with activated ß-catenin signalling. In addition, increasing HNF4α level by miR-34a inhibition slowed tumour progression of ß-catenin-activated HCC in mice. METHODS: We generated a Hnf4aflox/flox/ Apcflox/flox /TTR-CreERT2 (Hnf4a/Apc∆Hep ) mouse line and evaluated the impact of Hnf4a disruption on HCC development and liver homoeostasis. RESULTS: There was no significant impact of Hnf4a disruption on tumour onset and progression in Apc∆Hep model. However, we observed an unexpected phenotype in 28% of Hnf4a∆Hep mice maintained in a conventional animal facility, which presented disorganized portal triads, characterized by stenosis of the portal vein and increased number and size of hepatic arteries and bile ducts. These abnormal portal structures resemble the human idiopathic non-cirrhotic portal hypertension syndrome. We correlated the presence of portal remodelling with a higher expression of protein and mRNA levels of TGFß and BMP7, a key regulator of the TGFß-dependent endothelial-to-mesenchymal transition. CONCLUSION: These data demonstrate that HNF4α does not play a major role during ß-catenin-driven HCC, thus revealing that the tumour suppressor role of HNF4α is far more complex and dependent probably on its temporal expression and tumour context. However, HNF4α loss in adult hepatocytes could induce abnormal portal structures resembling the human idiopathic non-cirrhotic portal hypertension syndrome, which may result from endothelial- and epithelial-to-mesenchymal transitions.


Asunto(s)
Factor Nuclear 4 del Hepatocito/metabolismo , Hepatocitos/metabolismo , Neoplasias Hepáticas/genética , beta Catenina/metabolismo , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Proteína Morfogenética Ósea 7/metabolismo , Carcinogénesis , Diferenciación Celular , Línea Celular Tumoral , Transición Epitelial-Mesenquimal , Factor Nuclear 4 del Hepatocito/genética , Humanos , Neoplasias Hepáticas/metabolismo , Masculino , Ratones , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
13.
Front Immunol ; 9: 852, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29780382

RESUMEN

If misregulated, macrophage (Mϕ)-T cell interactions can drive chronic inflammation thereby causing diseases, such as rheumatoid arthritis (RA). We report that in a proinflammatory environment, granulocyte-Mϕ (GM-CSF)- and Mϕ colony-stimulating factor (M-CSF)-dependent Mϕs have dichotomous effects on T cell activity. While GM-CSF-dependent Mϕs show a highly stimulatory activity typical for M1 Mϕs, M-CSF-dependent Mϕs, marked by folate receptor ß (FRß), adopt an immunosuppressive M2 phenotype. We find the latter to be caused by the purinergic pathway that directs release of extracellular ATP and its conversion to immunosuppressive adenosine by co-expressed CD39 and CD73. Since we observed a misbalance between immunosuppressive and immunostimulatory Mϕs in human and murine arthritic joints, we devised a new strategy for RA treatment based on targeted delivery of a novel methotrexate (MTX) formulation to the immunosuppressive FRß+CD39+CD73+ Mϕs, which boosts adenosine production and curtails the dominance of proinflammatory Mϕs. In contrast to untargeted MTX, this approach leads to potent alleviation of inflammation in the murine arthritis model. In conclusion, we define the Mϕ extracellular purine metabolism as a novel checkpoint in Mϕ cell fate decision-making and an attractive target to control pathological Mϕs in immune-mediated diseases.


Asunto(s)
Artritis Reumatoide/inmunología , Diferenciación Celular , Macrófagos/inmunología , Macrófagos/metabolismo , Purinas/metabolismo , Adenosina/inmunología , Animales , Artritis Reumatoide/tratamiento farmacológico , Proliferación Celular , Modelos Animales de Enfermedad , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Humanos , Inmunosupresores/administración & dosificación , Inmunosupresores/uso terapéutico , Inflamación/tratamiento farmacológico , Factor Estimulante de Colonias de Macrófagos/farmacología , Masculino , Metotrexato/administración & dosificación , Metotrexato/uso terapéutico , Ratones , Monocitos/efectos de los fármacos , Líquido Sinovial/citología , Líquido Sinovial/inmunología
14.
Oncoimmunology ; 6(10): e1346765, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29123960

RESUMEN

Regressing tumors are usually associated with a large immune infiltrate, but the molecular and cellular interactions that govern a successful anti-tumor immunity remain elusive. Here, we have triggered type I Interferon (IFN) signaling in a breast tumor model (MMTV-PyMT) using 5,6-dimethylxanthenone-4-acetic acid (DMXAA), a ligand of the STimulator of Interferon Genes, STING. The 2 main events rapidly triggered by DMXAA in transplanted PyMT tumors are 1) the disruption of the tumor vasculature, followed by hypoxia and cell death; 2) the release of chemokines. Both events converged to trigger the recruitment of 2 waves of immune cells: a swift, massive recruitment of neutrophils, followed by a delayed rise in monocytes and CD8 T cells in the tumor mass. Depletion experiments in vivo revealed that myeloid cell subsets and T cells need to cooperate to achieve full-blown recruitment and activation at the tumor site and to induce effective secondary cell death leading to tumor regression (Illustration 1). Altogether, our study highlights that the tumor regression induced by the STING agonist DMXAA results from a cascade of events, with an initial vessel destruction followed by several infiltration waves of immune cells which have to cooperate to amplify and sustain the initial effect. We thus provide the first global and detailed kinetic analysis of the anti-tumoral effect of DMXAA and of its different articulated steps.

15.
Theranostics ; 7(11): 2757-2774, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28824714

RESUMEN

The growing field of cardio-oncology addresses the side effects of cancer treatment on the cardiovascular system. Here, we explored the cardiotoxicity of the antiangiogenic therapy, sunitinib, in the mouse heart from a diagnostic and therapeutic perspective. We showed that sunitinib induces an anaerobic switch of cellular metabolism within the myocardium which is associated with the development of myocardial fibrosis and reduced left ventricular ejection fraction as demonstrated by echocardiography. The capacity of positron emission tomography with [18F]fluorodeoxyglucose to detect the changes in cardiac metabolism caused by sunitinib was dependent on fasting status and duration of treatment. Pan proteomic analysis in the myocardium showed that sunitinib induced (i) an early metabolic switch with enhanced glycolysis and reduced oxidative phosphorylation, and (ii) a metabolic failure to use glucose as energy substrate, similar to the insulin resistance found in type 2 diabetes. Co-administration of the endothelin receptor antagonist, macitentan, to sunitinib-treated animals prevented both metabolic defects, restored glucose uptake and cardiac function, and prevented myocardial fibrosis. These results support the endothelin system in mediating the cardiotoxic effects of sunitinib and endothelin receptor antagonism as a potential therapeutic approach to prevent cardiotoxicity. Furthermore, metabolic and functional imaging can monitor the cardiotoxic effects and the benefits of endothelin antagonism in a theranostic approach.


Asunto(s)
Antineoplásicos/administración & dosificación , Antagonistas de los Receptores de Endotelina/metabolismo , Indoles/administración & dosificación , Miocardio/metabolismo , Pirroles/administración & dosificación , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Anaerobiosis , Animales , Antineoplásicos/efectos adversos , Glucólisis , Indoles/efectos adversos , Ratones Endogámicos C57BL , Miocardio/patología , Proteoma/análisis , Pirroles/efectos adversos , Sunitinib
16.
J Control Release ; 262: 170-181, 2017 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-28710005

RESUMEN

Despite the increasing number of clinical trials in gene therapy, no ideal methods still allow non-viral gene transfer in deep tissues such as the liver. We were interested in ultrasound (US)-mediated gene delivery to provide long term liver expression. For this purpose, new positively charged microbubbles were designed and complexed with pFAR4, a highly efficient small length miniplasmid DNA devoid of antibiotic resistance sequence. Sonoporation parameters, such as insonation time, acoustic pressure and duration of plasmid injection were controlled under ultrasound imaging guidance. The optimization of these various parameters was performed by bioluminescence optical imaging of luciferase reporter gene expression in the liver. Mice were injected with 50µg pFAR4-LUC either alone, or complexed with positively charged microbubbles, or co-injected with neutral MicroMarker™ microbubbles, followed by low ultrasound energy application to the liver. Injection of the pFAR4 encoding luciferase alone led to a transient transgene expression that lasted only for two days. The significant luciferase signal obtained with neutral microbubbles decreased over 2days and reached a plateau with a level around 1 log above the signal obtained with pFAR4 alone. With the newly designed positively charged microbubbles, we obtained a much stronger bioluminescence signal which increased over 2days. The 12-fold difference (p<0.05) between MicroMarker™ and our positively charged microbubbles was maintained over a period of 6months. Noteworthy, the positively charged microbubbles led to an improvement of 180-fold (p<0.001) as regard to free pDNA using unfocused ultrasound performed at clinically tolerated ultrasound amplitude. Transient liver damage was observed when using the cationic microbubble-pFAR4 complexes and the optimized sonoporation parameters. Immunohistochemistry analyses were performed to determine the nature of cells transfected. The pFAR4 miniplasmid complexed with cationic microbubbles allowed to transfect mostly hepatocytes compared to its co-injection with MicroMarker™ which transfected more preferentially endothelial cells.


Asunto(s)
ADN/administración & dosificación , Hígado/metabolismo , Microburbujas , Ondas Ultrasónicas , Animales , Técnicas de Transferencia de Gen , Células HeLa , Humanos , Lípidos/química , Hígado/diagnóstico por imagen , Luciferasas/genética , Luciferasas/metabolismo , Ratones Endogámicos BALB C , Plásmidos , Transgenes , Ultrasonografía
17.
J Mater Sci Mater Med ; 28(2): 32, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28108960

RESUMEN

Off the shelf scaffolds for replacing ultra-small diameter vascular grafts are valuable for reconstruction of diseased or damaged vessels. The limitations for such grafts include optimal handling with ready availability of varied lengths of grafts, graft patency with the ability to replace the function of active cellular mechanisms and adequate mechanical properties to maintain physicochemical function. We used a well-established, solvent casting method for potential tissue replacement scaffold fabrication with incorporated bioactive molecules, which we have previously explored to confer haemocompatibility. These grafts were tested in-vivo within the abdominal aorta of 10 Wistar rats and the patency was clinically and echographically evaluated. Haemocompatibility and endothelialisation were assessed on explants. Biofunctionalised scaffolds were also grafted subcutaneously and intraperitoneally to evaluate integration, inflammation and angiogenesis reactions. The potential wider applications of this dual acting scaffold were evaluated for its interactions with human dermal fibroblasts as well as bronchial epithelial cells. Physicochemical property evaluation of the functionalised grafts has clarified the mechanical strength and permeability. This study confirmed the microsurgical suturability of tubular grafts and graft patency of functionalized scaffolds. The study demonstrated the potential of a dual acting biofunctionalised scaffold's use for a wide range of tissue engineering applications where micro-porous, yet impermeable scaffolds are needed.


Asunto(s)
Aorta Abdominal/patología , Medicina Regenerativa/métodos , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Animales , Prótesis Vascular , Bronquios/citología , Células Epiteliales/citología , Diseño de Equipo , Fibroblastos/metabolismo , Humanos , Inflamación , Microscopía Electrónica de Rastreo , Microscopía Fluorescente , Permeabilidad , Polímeros/química , Presión , Ratas , Ratas Wistar , Piel/metabolismo , Estrés Mecánico , Temperatura , Resistencia a la Tracción
18.
Theranostics ; 7(2): 329-343, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28042338

RESUMEN

Tumor stiffening, stemming from aberrant production and organization of extracellular matrix (ECM), has been considered a predictive marker of tumor malignancy, non-invasively assessed by ultrasound shear wave elastography (SWE). Being more than a passive marker, tumor stiffening restricts the delivery of diagnostic and therapeutic agents to the tumor and per se could modulate cellular mechano-signaling, tissue inflammation and tumor progression. Current strategies to modify the tumor extracellular matrix are based on ECM-targeting chemical agents but also showed deleterious systemic effects. On-demand excitable nanomaterials have shown their ability to perturb the tumor microenvironment in a spatiotemporal-controlled manner and synergistically with chemotherapy. Here, we investigated the evolution of tumor stiffness as well as tumor integrity and progression, under the effect of mild hyperthermia and thermal ablation generated by light-exposed multi-walled carbon nanotubes (MWCNTs) in an epidermoid carcinoma mouse xenograft. SWE was used for real-time mapping of the tumor stiffness, both during the two near infrared irradiation sessions and over the days after the treatment. We observed a transient and reversible stiffening of the tumor tissue during laser irradiation, which was lowered at the second session of mild hyperthermia or photoablation. In contrast, over the days following photothermal treatment, the treated tumors exhibited a significant softening together with volume reduction, whereas non-treated growing tumors showed an increase of tumor rigidity. The organization of the collagen matrix and the distribution of CNTs revealed a spatio-temporal correlation between the presence of nanoheaters and the damages on collagen and cells. This study highlights nanohyperthermia as a promising adjuvant strategy to reverse tumor stiffening and normalize the mechanical tumor environment.


Asunto(s)
Antineoplásicos/administración & dosificación , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/terapia , Hipertermia Inducida/métodos , Nanoestructuras/administración & dosificación , Fotoquimioterapia/métodos , Animales , Modelos Animales de Enfermedad , Diagnóstico por Imagen de Elasticidad , Xenoinjertos , Rayos Láser , Ratones , Nanotubos de Carbono
19.
Gut ; 65(6): 1024-34, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-25792709

RESUMEN

OBJECTIVE: Hepatocellular carcinoma (HCC) is the most prevalent primary tumour of the liver. About a third of these tumours presents activating mutations of the ß-catenin gene. The molecular pathogenesis of HCC has been elucidated, but mortality remains high, and new therapeutic approaches, including treatments based on microRNAs, are required. We aimed to identify candidate microRNAs, regulated by ß-catenin, potentially involved in liver tumorigenesis. DESIGN: We used a mouse model, in which ß-catenin signalling was overactivated exclusively in the liver by the tamoxifen-inducible and Cre-Lox-mediated inactivation of the Apc gene. This model develops tumours with properties similar to human HCC. RESULTS: We found that miR-34a was regulated by ß-catenin, and significantly induced by the overactivation of ß-catenin signalling in mouse tumours and in patients with HCC. An inhibitor of miR-34a (locked nucleic acid, LNA-34a) exerted antiproliferative activity in primary cultures of hepatocyte. This inhibition of proliferation was associated with a decrease in cyclin D1 levels, orchestrated principally by HNF-4α, a target of miR-34a considered to act as a tumour suppressor in the liver. In vivo, LNA-34a approximately halved progression rates for tumours displaying ß-catenin activation together with an activation of caspases 2 and 3. CONCLUSIONS: This work demonstrates the key oncogenic role of miR-34a in liver tumours with ß-catenin gene mutations. We suggest that patients diagnosed with HCC with ß-catenin mutations could be treated with an inhibitor of miR-34a. The potential value of this strategy lies in the modulation of the tumour suppressor HNF-4α, which targets cyclin D1, and the induction of a proapoptotic programme.


Asunto(s)
Ciclina D1/genética , Neoplasias Hepáticas Experimentales/genética , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Mutación , beta Catenina/genética , Animales , Carcinoma Hepatocelular/terapia , Humanos , Neoplasias Hepáticas/terapia , Neoplasias Hepáticas Experimentales/terapia , Ratones
20.
J Biomed Nanotechnol ; 11(12): 2243-52, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26510317

RESUMEN

Methotrexate is the first line of treatment of rheumatoid arthritis. Since many patients become unresponsive to methotrexate treatment, only very expensive biological therapies are effective and increased methotrexate tolerance strategies need to be identified. Here we propose the encapsulation of methotrexate in a new liposomal formulation using a hydrophobic fragment of surfactant protein conjugated to a linker and folate to enhance their tolerance and efficacy. In this study we aim to evaluate the efficiency of this system to treat rheumatoid arthritis, by targeting folate receptor ß present at the surface of activated macrophages, key effector cells in this pathology. The specificity of our liposomal formulation to target folate receptor ß was investigated both in vitro as in vivo using a mouse model of arthritis (collagen-induced arthritis in DBA/1J mice strain). In both systems, the liposomal constructs were shown to be highly specific and efficient in targeting folate receptor ß. These liposomal formulations also significantly increase the clinical benefit of the encapsulated methotrexate in vivo in arthritic mice, together with reduced expression of CD39 and CD73 ectonucleotidases by joint-infiltrating macrophages. Thus, our formulation might be a promising cost effective way to treat rheumatoid arthritis and delay or reduce methotrexate intolerance.


Asunto(s)
Artritis Reumatoide/tratamiento farmacológico , Ácido Fólico/química , Metotrexato/efectos adversos , Metotrexato/farmacología , Animales , Línea Celular , Receptores de Folato Anclados a GPI/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Liposomas , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Metotrexato/administración & dosificación , Metotrexato/uso terapéutico , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA