Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Sci Rep ; 13(1): 16443, 2023 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-37777587

RESUMEN

Neuroblastoma, the most common type of pediatric extracranial solid tumor, causes 10% of childhood cancer deaths. Despite intensive multimodal treatment, the outcomes of high-risk neuroblastoma remain poor. We urgently need to develop new therapies with safe long-term toxicity profiles for rapid testing in clinical trials. Drug repurposing is a promising approach to meet these needs. Here, we investigated disulfiram, a safe and successful chronic alcoholism treatment with known anticancer and epigenetic effects. Disulfiram efficiently induced cell cycle arrest and decreased the viability of six human neuroblastoma cell lines at half-maximal inhibitory concentrations up to 20 times lower than its peak clinical plasma level in patients treated for chronic alcoholism. Disulfiram shifted neuroblastoma transcriptome, decreasing MYCN levels and activating neuronal differentiation. Consistently, disulfiram significantly reduced the protein level of lysine acetyltransferase 2A (KAT2A), drastically reducing acetylation of its target residues on histone H3. To investigate disulfiram's anticancer effects in an in vivo model of high-risk neuroblastoma, we developed a disulfiram-loaded emulsion to deliver the highly liposoluble drug. Treatment with the emulsion significantly delayed neuroblastoma progression in mice. These results identify KAT2A as a novel target of disulfiram, which directly impacts neuroblastoma epigenetics and is a promising candidate for repurposing to treat pediatric neuroblastoma.


Asunto(s)
Disulfiram , Neuroblastoma , Animales , Niño , Humanos , Ratones , Disuasivos de Alcohol/farmacología , Disuasivos de Alcohol/uso terapéutico , Línea Celular Tumoral , Disulfiram/farmacología , Disulfiram/uso terapéutico , Regulación hacia Abajo , Reposicionamiento de Medicamentos , Emulsiones/uso terapéutico , Histona Acetiltransferasas/efectos de los fármacos , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/genética
2.
Front Immunol ; 14: 1231916, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37675109

RESUMEN

Introduction: Natural Killer (NK) cells hold the potential to shift cell therapy from a complex autologous option to a universal off-the-shelf one. Although NK cells have demonstrated efficacy and safety in the treatment of leukemia, the limited efficacy of NK cell-based immunotherapies against solid tumors still represents a major hurdle. In the immunosuppressive tumor microenvironment (TME), inhibitory interactions between cancer and immune cells impair antitumoral immunity. KLRC1 gene encodes the NK cell inhibitory receptor NKG2A, which is a potent NK cell immune checkpoint. NKG2A specifically binds HLA-E, a non-classical HLA class I molecule frequently overexpressed in tumors, leading to the transmission of inhibitory signals that strongly impair NK cell function. Methods: To restore NK cell cytotoxicity against HLA-E+ tumors, we have targeted the NKG2A/HLA-E immune checkpoint by using a CRISPR-mediated KLRC1 gene editing. Results: KLRC1 knockout resulted in a reduction of 81% of NKG2A+ cell frequency in ex vivo expanded human NK cells post-cell sorting. In vitro, the overexpression of HLA-E by tumor cells significantly inhibited wild-type (WT) NK cell cytotoxicity with p-values ranging from 0.0071 to 0.0473 depending on tumor cell lines. In contrast, KLRC1 KO NK cells exhibited significantly higher cytotoxicity when compared to WT NK cells against four different HLA-E+ solid tumor cell lines, with p-values ranging from<0.0001 to 0.0154. Interestingly, a proportion of 43.5% to 60.2% of NKG2A- NK cells within the edited NK cell population was sufficient to reverse at its maximum the HLA-E-mediated inhibition of NK cell cytotoxicity. The expression of the activating receptor NKG2C was increased in KLRC1 KO NK cells and contributed to the improved NK cell cytotoxicity against HLA-E+ tumors. In vivo, the adoptive transfer of human KLRC1 KO NK cells significantly delayed tumor progression and increased survival in a xenogeneic mouse model of HLA-E+ metastatic breast cancer, as compared to WT NK cells (p = 0.0015). Conclusions: Our results demonstrate that KLRC1 knockout is an effective strategy to improve NK cell antitumor activity against HLA-E+ tumors and could be applied in the development of NK cell therapy for solid tumors.


Asunto(s)
Células Asesinas Naturales , Leucemia , Humanos , Animales , Ratones , Receptores de Células Asesinas Naturales , Transporte de Proteínas , Microambiente Tumoral , Antígenos HLA-E
3.
Stem Cell Reports ; 18(2): 597-612, 2023 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-36736326

RESUMEN

Humanization of mice with functional T cells currently relies on co-implantation of hematopoietic stem cells from fetal liver and autologous fetal thymic tissue (so-called BLT mouse model). Here, we show that NOD/SCID/IL2rγnull mice humanized with cord blood- derived CD34+ cells and implanted with allogeneic pediatric thymic tissues excised during cardiac surgeries (CCST) represent an alternative to BLT mice. CCST mice displayed a strong immune reconstitution, with functional T cells originating from CD34+ progenitor cells. They were equally susceptible to mucosal or intraperitoneal HIV infection and had significantly higher HIV-specific T cell responses. Antiretroviral therapy (ART) robustly suppressed viremia and reduced the frequencies of cells carrying integrated HIV DNA. As in BLT mice, we observed a complete viral rebound following ART interruption, suggesting the presence of HIV reservoirs. In conclusion, CCST mice represent a practical alternative to BLT mice, broadening the use of humanized mice for research.


Asunto(s)
Infecciones por VIH , Humanos , Ratones , Animales , Niño , Ratones SCID , Ratones Endogámicos NOD , Linfocitos T , Timo , Modelos Animales de Enfermedad , Ratones Noqueados
4.
Haematologica ; 108(2): 382-393, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36134452

RESUMEN

Acute lymphoblastic leukemia (ALL) is the most frequent cancer diagnosed in children. Despite the great progress achieved over the last 40 years, with cure rates now exceeding 85%, refractory or relapsed ALL still exhibit a dismal prognosis. This poor outcome reflects the lack of treatment options specifically targeting relapsed or refractory ALL. In order to address this gap, we performed whole-genome CRISPR/Cas drop-out screens on a panel of seven B-ALL cell lines. Our results demonstrate that while there was a significant overlap in gene essentiality between ALL cell lines and other cancer types survival of ALL cell lines was dependent on several unique metabolic pathways, including an exquisite sensitivity to GPX4 depletion and ferroptosis induction. Detailed molecular analysis of B-ALL cells suggest that they are primed to undergo ferroptosis as they exhibit high steady-state oxidative stress potential, a low buffering capacity, and a disabled GPX4-independent secondary lipid peroxidation detoxification pathway. Finally, we validated the sensitivity of BALL to ferroptosis induction using patient-derived B-ALL samples.


Asunto(s)
Ferroptosis , Leucemia-Linfoma Linfoblástico de Células Precursoras , Niño , Humanos , Fosfolípido Hidroperóxido Glutatión Peroxidasa/genética , Ferroptosis/genética , Línea Celular , Peroxidación de Lípido , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico
5.
Semin Hematol ; 57(4): 201-212, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33256913

RESUMEN

Cancer immunotherapy using genetically modified immune cells such as those expressing chimeric antigen receptors has shown dramatic outcomes in patients with refractory and relapsed malignancies. Natural killer (NK) cells as a member of the innate immune system, possessing both anticancer (cytotoxic) and proinflammatory (cytokine) responses to cancers and rare off-target toxicities have great potential for a wide range of cancer therapeutic settings. Therefore, improving NK cell antitumor activity through genetic modification is of high interest in the field of cancer immunotherapy. However, gene manipulation in primary NK cells has been challenging because of broad resistance to many genetic modification methods that work well in T cells. Here we review recent successful approaches for genetic and epigenetic modification of NK cells including epigenetic remodeling, transposons, mRNA-mediated gene delivery, lentiviruses, and CRISPR gene targeting.


Asunto(s)
Epigénesis Genética/genética , Terapia Genética/métodos , Inmunoterapia/métodos , Células Asesinas Naturales/inmunología , Neoplasias/terapia , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Neoplasias/inmunología
6.
Liver Int ; 35(1): 275-84, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24517723

RESUMEN

BACKGROUND: Some patients with autoimmune hepatitis (AIH), despite appropriate treatment, progress towards cirrhosis and liver failure, requiring transplantation. New biological agents targeting immune cell subtypes have been developed, with better specificity and longer-lasting effects than conventional wide-spectrum immunosuppressive drugs. AIMS: The goal of this study was to evaluate the effectiveness of low dose of αCD3 targeting therapy in a model of type 2 AIH. METHODS: This experimental model is based on xenoimmunization of C57BL/6 mice with DNA coding for human liver autoantigens. Mice with AIH were treated with five daily injections of low dose of αCD3 monoclonal antibody, before disease onset (5.5 months post-xenoimmunization) or during AIH (7 months post-xenoimmunization). Along with serum aminotransferases, autoantibody levels and end-point liver histology, spleen and liver-infiltrating lymphocytes were phenotyped by flow cytometry and immune response measured by lymphoproliferative assays. RESULTS: Before onset of AIH, treatment prevented the development of liver inflammation and tissue injury. During active AIH, low dose of αCD3 antibody therapy resulted in a resorption of liver inflammatory infiltrates, normalization of serum aminotransferas levels, reduced autoantibody titres, increased regulatory T cells and lowered proliferation of autoreactive liver lymphocytes. CONCLUSIONS: We report that low dose αCD3 antibody administration is an effective treatment for AIH in an experimental model of type 2 AIH. These data suggest that αCD3 antibody therapy could be tested in clinical trials as a rescue therapy for patients with uncontrolled AIH.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Complejo CD3/inmunología , Hepatitis Autoinmune/tratamiento farmacológico , Animales , Anticuerpos Monoclonales/farmacología , Autoantígenos/administración & dosificación , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Inducción de Remisión/métodos , Transaminasas/sangre
8.
J Autoimmun ; 42: 19-28, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23137675

RESUMEN

CD8(+) T-cell immune response to liver antigens is often functionally diminished or absent. This may occur via deletion of these autoaggressive T-cells, through the acquisition of an anergic phenotype, or via active suppression mediated by other cell populations. We generated a double transgenic model in which mice express CD8(+) T-cells specific for the lymphocytic choriomeningitis virus nucleoprotein (LCMV-NP) and LCMV-NP as a hepatic neo-autoantigen, to study the immunological response of potentially liver antigen autoaggressive CD8(+) T-cells. Autoreactive transgenic CD8(+) T-cells were analyzed for functionality and cytotoxic effector status. Despite severe peripheral deletion of liver-specific CD8(+) T-cells, a fraction of autoreactive NP-specific CD8(+) T-cells accumulate in liver, resulting in hepatocyte injury and production of auto-antibodies in both male and female mice. NP-specific intrahepatic T-cells showed capacity to proliferate, produce cytokines and up-regulate activation markers. These data provide in vivo evidence that autoreactive CD8(+) T-cells are activated in the liver and developed an inflammatory process, but require additional factors to cause severe autoimmune destruction of hepatocytes. Our new model will provide a valuable tool for further exploration of the immunological response involved in inflammatory liver diseases, including autoimmune hepatitis.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Hepatocitos/inmunología , Hígado/inmunología , Nucleoproteínas/metabolismo , Animales , Antígenos Virales/genética , Antígenos Virales/inmunología , Apoptosis/genética , Apoptosis/inmunología , Células Cultivadas , Citotoxicidad Inmunológica/genética , Modelos Animales de Enfermedad , Femenino , Hepatitis Autoinmune/inmunología , Hepatocitos/patología , Virus de la Coriomeningitis Linfocítica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Nucleoproteínas/genética , Nucleoproteínas/inmunología , Especificidad de Órganos/genética , Especificidad de Órganos/inmunología , Transgenes/genética
9.
PLoS One ; 7(10): e48192, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23110209

RESUMEN

The liver must keep equilibrium between immune tolerance and immunity in order to protect itself from pathogens while maintaining tolerance to food antigens. An imbalance between these two states could result in an inflammatory liver disease. The aims of this study were to identify factors responsible for a break of tolerance and characterize the subsequent restoration of liver immune homeostasis. A pro-inflammatory environment was created in the liver by the co-administration of TLR ligands CpG and Poly(I:C) in presence or absence of activated liver-specific autoreactive CD8(+) T cells. Regardless of autoreactive CD8(+) T cells, mice injected with CpG and Poly(I:C) showed elevated serum ALT levels and a transient liver inflammation. Both CpG/Poly(I:C) and autoreactive CD8(+)T cells induced expression of TLR9 and INF-γ by the liver, and an up-regulation of homing and adhesion molecules CXCL9, CXCL10, CXCL16, ICAM-1 and VCAM-1. Transferred CFSE-labeled autoreactive CD8(+) T cells, in presence of TLR3 and 9 ligands, were recruited by the liver and spleen and proliferated. This population then contracted by apoptosis through intrinsic and extrinsic pathways. Up-regulation of FasL and PD-L1 in the liver was observed. In conclusion, TLR-mediated activation of the innate immune system results in a pro-inflammatory environment that promotes the recruitment of lymphocytes resulting in bystander hepatitis. Despite this pro-inflammatory environment, the presence of autoreactive CD8(+) T cells is not sufficient to sustain an autoimmune response against the liver and immune homeostasis is rapidly restored through the apoptosis of T cells.


Asunto(s)
Inflamación/inmunología , Inflamación/metabolismo , Hígado/inmunología , Hígado/metabolismo , Linfocitos T/metabolismo , Animales , Antígeno B7-H1/metabolismo , Western Blotting , Antígenos CD13/sangre , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Quimiocina CXCL10/metabolismo , Quimiocina CXCL16 , Quimiocina CXCL6/metabolismo , Quimiocina CXCL9/metabolismo , Proteína Ligando Fas/metabolismo , Citometría de Flujo , Homeostasis/fisiología , Inmunidad Innata , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Molécula 1 de Adhesión Intercelular/metabolismo , Ratones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/inmunología , Molécula 1 de Adhesión Celular Vascular/metabolismo
10.
J Pediatr ; 160(5): 871-4.e1, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22341950
11.
Liver Int ; 31(9): 1306-14, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22093453

RESUMEN

BACKGROUND: Epidemiological and laboratory evidences led to the hypothesis that molecular mimicry between viruses and self-proteins could be linked to the onset of autoimmune hepatitis (AIH). Hepatotropic viruses could be good candidates, as a pro-inflammatory environment may facilitate the development of AIH. AIMS: The aims of this study were to test a virus ability to induce an AIH through molecular mimicry and the influence of hepatic inflammation in this process. METHODS: C57BL/6 mice were injected i.v. or i.m. with recombinant adenoviral vectors (RecAdV) encoding for human type 2 AIH antigens to target xenoantigens expression in the liver and to create a transient hepatitis (i.v.) or for 'peripheral' xenoantigens expression (i.m.). Liver injury and B-cell response were evaluated. RESULTS: Late-onset hepatitis was observed 8 months after i.v. or i.m. RecAdV injections, despite presence or absence of an initial transient hepatitis. Intensity of B-cell response was similar for both type of injections, but the Ig isotypes produced were different. B-cell autoimmune response spread to several liver proteins. CONCLUSIONS: Liver autoimmune response can be initiated using molecular mimicry over a long period of time, validating the hit-and-run hypothesis. Initial liver inflammatory injury is neither necessary, nor detrimental to the development of AIH. These results highlight the significance of initial events on the pathogenesis of autoimmune liver injury.


Asunto(s)
Antígenos Heterófilos/inmunología , Hepatitis Autoinmune/inmunología , Hígado/inmunología , Imitación Molecular , Adenoviridae/genética , Amoníaco-Liasas/genética , Amoníaco-Liasas/inmunología , Animales , Antígenos Heterófilos/biosíntesis , Antígenos Heterófilos/genética , Linfocitos B/inmunología , Linfocitos B/virología , Antígeno CTLA-4/genética , Antígeno CTLA-4/inmunología , Citocromo P-450 CYP2D6/genética , Citocromo P-450 CYP2D6/inmunología , Femenino , Técnicas de Transferencia de Gen , Vectores Genéticos , Glutamato Formimidoiltransferasa/genética , Glutamato Formimidoiltransferasa/inmunología , Hepatitis Autoinmune/patología , Hepatitis Autoinmune/virología , Humanos , Inyecciones Intramusculares , Inyecciones Intravenosas , Hígado/patología , Hígado/virología , Ratones , Ratones Endogámicos C57BL , Enzimas Multifuncionales , Células TH1/inmunología , Células TH1/virología , Células Th2/inmunología , Células Th2/virología , Factores de Tiempo
12.
J Autoimmun ; 26(2): 82-9, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16380229

RESUMEN

Genetic predisposition is recognized as an important factor for the development of autoimmune hepatitis (AIH). To assess the potential contribution of MHC and non-MHC genes, type 2 AIH was reproduced in three mice strains, taking advantage of their different genetic makeup with regard to MHC and non-MHC genes. Mice (C57BL/6, 129/Sv and BALB/c) were DNA vaccinated with a pCMV-CTLA4-CYP2D6-FTCD plasmid coding for the extracellular region of CTLA-4 and for the antigenic region of the CYP2D6 and FTCD, and with pCMV-IL12. ALT and total IgG levels, liver histology, FACS analysis and liver T-cell cytotoxicity assays were monitored up to 8 months post-injection. C57BL/6 mice showed elevated serum ALT levels, autoantibodies, antigen-specific cytotoxic T-cells and lobular and periportal inflammatory infiltrate. The 129/Sv mice showed slightly elevated ALT levels, sparse liver lobular infiltrate and cytotoxic T-cells. The BALB/c mice showed no liver inflammation. All mice had elevated total serum IgG levels. This murine model of type 2 AIH shows that MHC and non-MHC genes contribute to the susceptibility to autoimmune hepatitis. The understanding of the genetic determinants implicated in AIH development will be a major advance in the study of its pathogenesis and could lead to a better diagnostic approach and preventive strategies.


Asunto(s)
Predisposición Genética a la Enfermedad , Hepatitis Autoinmune/genética , Complejo Mayor de Histocompatibilidad/genética , Alanina Transaminasa/sangre , Amoníaco-Liasas , Animales , Antígenos CD , Antígenos de Diferenciación/análisis , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/inmunología , Autoanticuerpos , Linfocitos B/inmunología , Antígeno CTLA-4 , Citocromo P-450 CYP2D6/genética , Citocromo P-450 CYP2D6/inmunología , Modelos Animales de Enfermedad , Glutamato Formimidoiltransferasa , Hepatitis Autoinmune/inmunología , Hepatitis Autoinmune/patología , Inmunoglobulina G/sangre , Ratones , Ratones Endogámicos , Complejos Multienzimáticos/genética , Complejos Multienzimáticos/inmunología , Enzimas Multifuncionales , Plásmidos/genética , Plásmidos/inmunología , Linfocitos T Citotóxicos/inmunología , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA