Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-38328145

RESUMEN

Xenografting human cancer tissues into mice to test new cures against cancers is critical for understanding and treating the disease. However, only a few inbred strains of mice are used to study cancers, and derivatives of mainly one strain, mostly NOD/ShiLtJ, are used for therapy efficacy studies. As it has been demonstrated when human cancer cell lines or patient-derived tissues (PDX) are xenografted into mice, the neoplastic cells are human but the supporting cells that comprise the tumor (the stroma) are from the mouse. Therefore, results of studies of xenografted tissues are influenced by the host strain. We previously published that when the same neoplastic cells are xenografted into different mouse strains, the pattern of tumor growth, histology of the tumor, number of immune cells infiltrating the tumor, and types of circulating cytokines differ depending on the strain. Therefore, to better comprehend the behavior of cancer in vivo, one must xenograft multiple mouse strains. Here we describe and report a series of methods that we used to reveal the genes and proteins expressed when the same cancer cell line, MDA-MB-231, is xenografted in different hosts. First, using proteomic analysis, we show how to use the same cell line in vivo to reveal the protein changes in the neoplastic cell that help it adapt to its host. Then, we show how different hosts respond molecularly to the same cell line. We also find that using multiple strains can reveal a more suitable host than those traditionally used for a "difficult to xenograft" PDX. In addition, using complex trait genetics, we illustrate a feasible method for uncovering the alleles of the host that support tumor growth. Finally, we demonstrate that Diversity Outbred mice, the epitome of a model of mouse-strain genetic diversity, can be xenografted with human cell lines or PDX using 2-deoxy-D-glucose treatment.

2.
Aging Cell ; 22(4): e13792, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36840360

RESUMEN

Diverse mouse strains have different health and life spans, mimicking the diversity among humans. To capture conserved aging signatures, we studied long-lived C57BL/6J and short-lived NZO/HILtJ mouse strains by profiling transcriptomes and epigenomes of immune cells from peripheral blood and the spleen from young and old mice. Transcriptional activation of the AP-1 transcription factor complex, particularly Fos, Junb, and Jun genes, was the most significant and conserved aging signature across tissues and strains. ATAC-seq data analyses showed that the chromatin around these genes was more accessible with age and there were significantly more binding sites for these TFs with age across all studied tissues, targeting pro-inflammatory molecules including Il6. Age-related increases in binding sites of JUN and FOS factors were also conserved in human peripheral blood ATAC-seq data. Single-cell RNA-seq data from the mouse aging cell atlas Tabula Muris Senis showed that the expression of these genes increased with age in B, T, NK cells, and macrophages, with macrophages from old mice expressing these molecules more abundantly than other cells. Functional data showed that upon myeloid cell activation via poly(I:C), the levels of JUN protein and its binding activity increased more significantly in spleen cells from old compared to young mice. In addition, upon activation, old cells produced more IL6 compared to young cells. In sum, we showed that the aging-related transcriptional activation of Jun and Fos family members in AP-1 complex is conserved across immune tissues and long- and short-living mouse strains, possibly contributing to increased inflammation with age.


Asunto(s)
Proteínas Proto-Oncogénicas c-fos , Factor de Transcripción AP-1 , Animales , Humanos , Ratones , Envejecimiento/genética , Interleucina-6/metabolismo , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo , Activación Transcripcional
3.
Dis Model Mech ; 15(9)2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-36037073

RESUMEN

The lack of genetically diverse preclinical animal models in basic biology and efficacy testing has been cited as a potential cause of failure in clinical trials. We developed and characterized five diverse RAG1 null mouse strains as models that allow xenografts to grow. In these strains, we characterized the growth of breast cancer, leukemia and glioma cell lines. We found a wide range of growth characteristics that were far more dependent on strain than tumor type. For the breast cancer cell line, we characterized the spectrum of xenograft/tumor growth at structural, histological, cellular and molecular levels across each strain, and found that each strain captures unique structural components of the stroma. Furthermore, we showed that the increase in tumor-infiltrating myeloid CD45+ cells and the amount of circulating cytokine IL-6 and chemokine KC (also known as CXCL1) is associated with a higher tumor size in different strains. This resource is available to study established human xenografts, as well as difficult-to-xenograft tumors and growth of hematopoietic stems cells, and to decipher the role of myeloid cells in the development of spontaneous cancers.


Asunto(s)
Neoplasias de la Mama , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Xenoinjertos , Humanos , Ratones , Ratones Noqueados , Trasplante Heterólogo , Ensayos Antitumor por Modelo de Xenoinjerto
4.
J Cell Mol Med ; 25(1): 229-243, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33249764

RESUMEN

Heart failure is the common final pathway of several cardiovascular conditions and a major cause of morbidity and mortality worldwide. Aberrant activation of the adaptive immune system in response to myocardial necrosis has recently been implicated in the development of heart failure. The ß-adrenergic agonist isoproterenol hydrochloride is used for its cardiac effects in a variety of different dosing regimens with high doses causing acute cardiomyocyte necrosis. To assess whether isoproterenol-induced cardiomyocyte necrosis triggers an adaptive immune response against the heart, we treated C57BL/6J mice with a single intraperitoneal injection of isoproterenol. We confirmed tissue damage reminiscent of human type 2 myocardial infarction. This is followed by an adaptive immune response targeting the heart as demonstrated by the activation of T cells, the presence of anti-heart auto-antibodies in the serum as late as 12 weeks after initial challenge and IgG deposition in the myocardium. All of these are hallmark signs of an established autoimmune response. Adoptive transfer of splenocytes from isoproterenol-treated mice induces left ventricular dilation and impairs cardiac function in healthy recipients. In summary, a single administration of a high dose of isoproterenol is a suitable high-throughput model for future studies of the pathological mechanisms of anti-heart autoimmunity and to test potential immunomodulatory therapeutic approaches.


Asunto(s)
Inmunidad Adaptativa , Infarto del Miocardio/inmunología , Miocardio/patología , Traslado Adoptivo , Animales , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Femenino , Fibrosis , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Isoproterenol , Antígenos Comunes de Leucocito/metabolismo , Masculino , Ratones Endogámicos C57BL , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Necrosis , Especificidad de Órganos , Bazo/inmunología , Sístole , Linfocitos T Colaboradores-Inductores/inmunología , Vasodilatación
5.
Circulation ; 143(8): 821-836, 2021 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-33297741

RESUMEN

BACKGROUND: Ischemic heart disease is a leading cause of heart failure and despite advanced therapeutic options, morbidity and mortality rates remain high. Although acute inflammation in response to myocardial cell death has been extensively studied, subsequent adaptive immune activity and anti-heart autoimmunity may also contribute to the development of heart failure. After ischemic injury to the myocardium, dendritic cells (DC) respond to cardiomyocyte necrosis, present cardiac antigen to T cells, and potentially initiate a persistent autoimmune response against the heart. Cross-priming DC have the ability to activate both CD4+ helper and CD8+ cytotoxic T cells in response to necrotic cells and may thus be crucial players in exacerbating autoimmunity targeting the heart. This study investigates a role for cross-priming DC in post-myocardial infarction immunopathology through presentation of self-antigen from necrotic cardiac cells to cytotoxic CD8+ T cells. METHODS: We induced type 2 myocardial infarction-like ischemic injury in the heart by treatment with a single high dose of the ß-adrenergic agonist isoproterenol. We characterized the DC population in the heart and mediastinal lymph nodes and analyzed long-term cardiac immunopathology and functional decline in wild type and Clec9a-depleted mice lacking DC cross-priming function. RESULTS: A diverse DC population, including cross-priming DC, is present in the heart and activated after ischemic injury. Clec9a-/- mice deficient in DC cross-priming are protected from persistent immune-mediated myocardial damage and decline of cardiac function, likely because of dampened activation of cytotoxic CD8+ T cells. CONCLUSION: Activation of cytotoxic CD8+ T cells by cross-priming DC contributes to exacerbation of postischemic inflammatory damage of the myocardium and corresponding decline in cardiac function. Importantly, this provides novel therapeutic targets to prevent postischemic immunopathology and heart failure.


Asunto(s)
Reactividad Cruzada , Células Dendríticas/inmunología , Miocardio/patología , Animales , Presentación de Antígeno , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Femenino , Insuficiencia Cardíaca/patología , Humanos , Lectinas Tipo C/deficiencia , Lectinas Tipo C/genética , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Infarto del Miocardio/inmunología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocardio/inmunología , Miocardio/metabolismo , Receptores de Quimiocina/metabolismo , Receptores Inmunológicos/deficiencia , Receptores Inmunológicos/genética
6.
Genes Cancer ; 11(1-2): 83-94, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32577159

RESUMEN

Known as the guardian of the genome, transformation-related protein 53 (TRP53) is a well -known tumor suppressor. Here, we describe a novel TRP53 deficient mouse model on a tumor prone background-SJL/J mice. The absence of TRP53 (TRP53 nullizygosity) leads to a shift in the tumor spectrum from a non-Hodgkin's-like disease to thymic lymphomas and testicular teratomas at a very rapid tumor onset averaging ~12 weeks of age. In haplotype studies, comparing tumor prone versus tumor resistant Trp53 null mouse strains, we found that other tumor suppressor, DNA repair and/or immune system genes modulate tumor incidence in TRP53 null strains, suggesting that even a strong tumor suppressor such as TRP53 is modulated by genetic background. Due to their rapid development of tumors, the SJL/J TRP53 null mice generated here can be used as an efficient chemotherapy or immunotherapy screening mouse model.

7.
J Immunol ; 204(5): 1091-1100, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31988182

RESUMEN

Almost a decade has passed since the approval of belimumab, an mAb directed against B lymphocyte stimulation and the first targeted therapy approved for systemic lupus erythematous (SLE) in over 50 y. Although well tolerated, the efficacy of belimumab remains limited and is not labeled for patients suffering from nephritis, the leading cause of patient mortality. We sought to explore alternative targets of autoreactive B lymphocytes through manipulation of affinity maturation. The BXSB/MpJ mouse, a well-established model of human SLE, develops elevated antinuclear Abs and immune complex-mediated nephritis along with other manifestations of SLE-like disease. To limit interfering with critical background genetics, we used CRISPR-Cas9 to disrupt activation-induced cytidine deaminase (AID; Aicda) directly in BXSB zygotes. Homozygous null mice demonstrated significantly prolonged survival compared with wild-type. Although mice continued to develop plasma cells, splenic follicular structure was restored, and renal pathology was reduced. Mice developed expanded germinal center B lymphocyte populations as in other models of AID deficiency as well as increased populations of CD73+ B lymphocytes. Treatment with the small molecule inhibitor of RAD51, 4,4'-diisothiocyano-2,2'-stilbenedisulfonic acid, resulted in minimal changes in disease markers in BXSB mice. The prolonged survival in AID-deficient BXSB mice appears attributed primarily to the reduced renal pathology, warranting further exploration, as current therapeutics targeting lupus nephritis are limited and, thus, in great demand.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Citidina Desaminasa/inmunología , Lupus Eritematoso Sistémico/inmunología , Animales , Subgrupos de Linfocitos B/patología , Sistemas CRISPR-Cas , Citidina Desaminasa/genética , Modelos Animales de Enfermedad , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/patología , Ratones , Ratones Noqueados
8.
Dis Model Mech ; 12(8)2019 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-31324689

RESUMEN

Hemorrhagic myocarditis is a potentially fatal complication of excessive levels of systemic inflammation. It has been reported in viral infection, but is also possible in systemic autoimmunity. Epicutaneous treatment of mice with the Toll-like receptor 7 (TLR-7) agonist Resiquimod induces auto-antibodies and systemic tissue damage, including in the heart, and is used as an inducible mouse model of systemic lupus erythematosus (SLE). Here, we show that overactivation of the TLR-7 pathway of viral recognition by Resiquimod treatment of CFN mice induces severe thrombocytopenia and internal bleeding, which manifests most prominently as hemorrhagic myocarditis. We optimized a cardiac magnetic resonance (CMR) tissue mapping approach for the in vivo detection of diffuse infiltration, fibrosis and hemorrhages using a combination of T1, T2 and T2* relaxation times, and compared results with ex vivo histopathology of cardiac sections corresponding to CMR tissue maps. This allowed detailed correlation between in vivo CMR parameters and ex vivo histopathology, and confirmed the need to include T2* measurements to detect tissue iron for accurate interpretation of pathology associated with CMR parameter changes. In summary, we provide detailed histological and in vivo imaging-based characterization of acute hemorrhagic myocarditis as an acute cardiac complication in the mouse model of Resiquimod-induced SLE, and a refined CMR protocol to allow non-invasive longitudinal in vivo studies of heart involvement in acute inflammation. We propose that adding T2* mapping to CMR protocols for myocarditis diagnosis improves diagnostic sensitivity and interpretation of disease mechanisms.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Hemorragia/diagnóstico por imagen , Imágenes de Resonancia Magnética Multiparamétrica , Miocarditis/diagnóstico por imagen , Receptor Toll-Like 7/agonistas , Animales , Femenino , Hemorragia/complicaciones , Hemorragia/patología , Humanos , Imidazoles , Inflamación/complicaciones , Inflamación/patología , Hierro/metabolismo , Masculino , Ratones Endogámicos C57BL , Miocarditis/complicaciones , Miocarditis/patología , Trombocitopenia/complicaciones , Trombocitopenia/patología
9.
Dis Model Mech ; 12(3)2019 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-30858306

RESUMEN

Patients suffering from systemic autoimmune diseases are at significant risk of cardiovascular complications. This can be due to systemically increased levels of inflammation leading to accelerated atherosclerosis, or due to direct damage to the tissues and cells of the heart. Cardiac complications include an increased risk of myocardial infarction, myocarditis and dilated cardiomyopathy, valve disease, endothelial dysfunction, excessive fibrosis, and bona fide autoimmune-mediated tissue damage by autoantibodies or auto-reactive cells. There is, however, still a considerable need to better understand how to diagnose and treat cardiac complications in autoimmune patients. A range of inducible and spontaneous mouse models of systemic autoimmune diseases is available for mechanistic and therapeutic studies. For this Review, we systematically collated information on the cardiac phenotype in the most common inducible, spontaneous and engineered mouse models of systemic lupus erythematosus, rheumatoid arthritis and systemic sclerosis. We also highlight selected lesser-known models of interest to provide researchers with a decision framework to choose the most suitable model for their study of heart involvement in systemic autoimmunity.


Asunto(s)
Autoinmunidad , Miocardio/patología , Animales , Modelos Animales de Enfermedad , Ratones , Fenotipo
10.
Cancer Biol Ther ; 20(2): 169-182, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30183475

RESUMEN

Targeting the early steps of the glycolysis pathway in cancers is a well-established therapeutic strategy; however, the doses required to elicit a therapeutic effect on the cancer can be toxic to the patient. Consequently, numerous preclinical and clinical studies have combined glycolytic blockade with other therapies. However, most of these other therapies do not specifically target cancer cells, and thus adversely affect normal tissue. Here we first show that a diverse number of cancer models - spontaneous, patient-derived xenografted tumor samples, and xenografted human cancer cells - can be efficiently targeted by 2-deoxy-D-Glucose (2DG), a well-known glycolytic inhibitor. Next, we tested the cancer-cell specificity of a therapeutic compound using the MEC1 cell line, a chronic lymphocytic leukemia (CLL) cell line that expresses activation induced cytidine deaminase (AID). We show that MEC1 cells, are susceptible to 4,4'-Diisothiocyano-2,2'-stilbenedisulfonic acid (DIDS), a specific RAD51 inhibitor. We then combine 2DG and DIDS, each at a lower dose and demonstrate that this combination is more efficacious than fludarabine, the current standard- of- care treatment for CLL. This suggests that the therapeutic blockade of glycolysis together with the therapeutic inhibition of RAD51-dependent homologous recombination can be a potentially beneficial combination for targeting AID positive cancer cells with minimal adverse effects on normal tissue. Implications: Combination therapy targeting glycolysis and specific RAD51 function shows increased efficacy as compared to standard of care treatments in leukemias.


Asunto(s)
Ácido 4,4'-Diisotiocianostilbeno-2,2'-Disulfónico/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Desoxiglucosa/farmacología , Neoplasias/tratamiento farmacológico , Recombinasa Rad51/antagonistas & inhibidores , Ácido 4,4'-Diisotiocianostilbeno-2,2'-Disulfónico/administración & dosificación , Animales , Línea Celular Tumoral , Desoxiglucosa/administración & dosificación , Sinergismo Farmacológico , Femenino , Glucólisis/efectos de los fármacos , Humanos , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Leucemia Linfocítica Crónica de Células B/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Neoplasias/metabolismo , Recombinasa Rad51/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Cardiovasc Res ; 114(11): 1445-1461, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-30010800

RESUMEN

Following a myocardial infarction (MI), the immune system helps to repair ischaemic damage and restore tissue integrity, but excessive inflammation has been implicated in adverse cardiac remodelling and development towards heart failure (HF). Pre-clinical studies suggest that timely resolution of inflammation may help prevent HF development and progression. Therapeutic attempts to prevent excessive post-MI inflammation in patients have included pharmacological interventions ranging from broad immunosuppression to immunomodulatory approaches targeting specific cell types or factors with the aim to maintain beneficial aspects of the early post-MI immune response. These include the blockade of early initiators of inflammation including reactive oxygen species and complement, inhibition of mast cell degranulation and leucocyte infiltration, blockade of inflammatory cytokines, and inhibition of adaptive B and T-lymphocytes. Herein, we provide a systematic review on post-MI immunomodulation trials and a meta-analysis of studies targeting the inflammatory cytokine Interleukin-1. Despite an enormous effort into a significant number of clinical trials on a variety of targets, a striking heterogeneity in study population, timing and type of treatment, and highly variable endpoints limits the possibility for meaningful meta-analyses. To conclude, we highlight critical considerations for future studies including (i) the therapeutic window of opportunity, (ii) immunological effects of routine post-MI medication, (iii) stratification of the highly diverse post-MI patient population, (iv) the potential benefits of combining immunomodulatory with regenerative therapies, and at last (v) the potential side effects of immunotherapies.


Asunto(s)
Antiinflamatorios/uso terapéutico , Insuficiencia Cardíaca/tratamiento farmacológico , Inmunosupresores/uso terapéutico , Mediadores de Inflamación/antagonistas & inhibidores , Interleucina-1/antagonistas & inhibidores , Infarto del Miocardio/tratamiento farmacológico , Antiinflamatorios/efectos adversos , Ensayos Clínicos como Asunto , Insuficiencia Cardíaca/inmunología , Insuficiencia Cardíaca/metabolismo , Humanos , Inmunosupresores/efectos adversos , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Interleucina-1/inmunología , Interleucina-1/metabolismo , Infarto del Miocardio/inmunología , Infarto del Miocardio/metabolismo , Factores de Riesgo , Transducción de Señal/efectos de los fármacos , Resultado del Tratamiento
12.
Adv Exp Med Biol ; 1003: 35-70, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28667553

RESUMEN

This chapter will discuss the role of cardiac fibroblasts as a target of various immunological inputs as well as an immunomodulatory hub of the heart through interaction with immune cell types and chemokine or cytokine signaling. While the purpose of this chapter is to explore the immunomodulatory properties of cardiac fibroblasts, it is important to note that cardiac fibroblasts are not a homogeneous cell type, but have a unique embryological origin and molecular identity. Specific properties of cardiac fibroblasts may influence the way they interact with the heart microenvironment to promote healthy homeostatic function or respond to pathological insults. Therefore, we will briefly discuss these aspects of cardiac fibroblast biology and then focus on their immunomodulatory role in the heart.


Asunto(s)
Inmunidad Adaptativa , Cardiomiopatías/inmunología , Fibroblastos/inmunología , Inmunidad Innata , Miocardio/inmunología , Animales , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Matriz Extracelular/inmunología , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis , Humanos , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Miocardio/metabolismo , Miocardio/patología , Fenotipo , Transducción de Señal
13.
J Immunol ; 198(11): 4255-4267, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28461573

RESUMEN

B lymphocytes play a key role in type 1 diabetes (T1D) development by serving as a subset of APCs preferentially supporting the expansion of autoreactive pathogenic T cells. As a result of their pathogenic importance, B lymphocyte-targeted therapies have received considerable interest as potential T1D interventions. Unfortunately, the B lymphocyte-directed T1D interventions tested to date failed to halt ß cell demise. IgG autoantibodies marking humans at future risk for T1D indicate that B lymphocytes producing them have undergone the affinity-maturation processes of class switch recombination and, possibly, somatic hypermutation. This study found that CRISPR/Cas9-mediated ablation of the activation-induced cytidine deaminase gene required for class switch recombination/somatic hypermutation induction inhibits T1D development in the NOD mouse model. The activation-induced cytidine deaminase protein induces genome-wide DNA breaks that, if not repaired through RAD51-mediated homologous recombination, result in B lymphocyte death. Treatment with the RAD51 inhibitor 4,4'-diisothiocyanatostilbene-2, 2'-disulfonic acid also strongly inhibited T1D development in NOD mice. The genetic and small molecule-targeting approaches expanded CD73+ B lymphocytes that exert regulatory activity suppressing diabetogenic T cell responses. Hence, an initial CRISPR/Cas9-mediated genetic modification approach has identified the AID/RAD51 axis as a target for a potentially clinically translatable pharmacological approach that can block T1D development by converting B lymphocytes to a disease-inhibitory CD73+ regulatory state.


Asunto(s)
Linfocitos B Reguladores/inmunología , Proteínas Portadoras/antagonistas & inhibidores , Citidina Desaminasa/antagonistas & inhibidores , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/prevención & control , Activación de Linfocitos , Proteínas Nucleares/antagonistas & inhibidores , Ácido 4,4'-Diisotiocianostilbeno-2,2'-Disulfónico/farmacología , 5'-Nucleotidasa/inmunología , Animales , Autoanticuerpos/inmunología , Sistemas CRISPR-Cas , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Citidina Desaminasa/genética , Citidina Desaminasa/metabolismo , Proteínas de Unión al ADN , Diabetes Mellitus Experimental , Cambio de Clase de Inmunoglobulina , Ratones , Ratones Endogámicos NOD , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas de Unión al ARN , Hipermutación Somática de Inmunoglobulina
14.
Dis Model Mech ; 10(3): 259-270, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28250051

RESUMEN

Systemic autoimmune diseases such as systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA) show significant heart involvement and cardiovascular morbidity, which can be due to systemically increased levels of inflammation or direct autoreactivity targeting cardiac tissue. Despite high clinical relevance, cardiac damage secondary to systemic autoimmunity lacks inducible rodent models. Here, we characterise immune-mediated cardiac tissue damage in a new model of SLE induced by topical application of the Toll-like receptor 7/8 (TLR7/8) agonist Resiquimod. We observe a cardiac phenotype reminiscent of autoimmune-mediated dilated cardiomyopathy, and identify auto-antibodies as major contributors to cardiac tissue damage. Resiquimod-induced heart disease is a highly relevant mouse model for mechanistic and therapeutic studies aiming to protect the heart during autoimmunity.


Asunto(s)
Autoinmunidad/efectos de los fármacos , Cardiomiopatía Dilatada/inducido químicamente , Imidazoles/efectos adversos , Miocarditis/inducido químicamente , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 8/agonistas , Inmunidad Adaptativa/efectos de los fármacos , Traslado Adoptivo , Animales , Autoanticuerpos/sangre , Cardiomiopatía Dilatada/complicaciones , Cardiomiopatía Dilatada/inmunología , Cardiomiopatía Dilatada/fisiopatología , Modelos Animales de Enfermedad , Femenino , Variación Genética , Pruebas de Función Cardíaca , Inmunidad Celular/efectos de los fármacos , Inflamación/patología , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/patología , Masculino , Mutación/genética , Miocarditis/complicaciones , Miocarditis/inmunología , Miocarditis/fisiopatología , Miocardio/patología , Bazo/patología , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 8/metabolismo
15.
J Exp Med ; 210(5): 1021-33, 2013 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-23589568

RESUMEN

Activation-induced cytidine deaminase (AID) is critical in normal B cells to initiate somatic hypermutation and immunoglobulin class switch recombination. Accumulating evidence suggests that AID is also prooncogenic, inducing cancer-promoting mutations or chromosome rearrangements. In this context, we find that AID is expressed in >40% of primary human chronic lymphocytic leukemia (CLL) cases, consistent with other reports. Using a combination of human B lymphoid leukemia cells and mouse models, we now show that AID expression can be harnessed for antileukemic effect, after inhibition of the RAD51 homologous recombination (HR) factor with 4,4'-diisothiocyanatostilbene-2-2'-disulfonic acid (DIDS). As a proof of principle, we show that DIDS treatment inhibits repair of AID-initiated DNA breaks, induces apoptosis, and promotes cytotoxicity preferentially in AID-expressing human CLL. This reveals a novel antineoplastic role of AID that can be triggered by inhibition of HR, suggesting a potential new paradigm to treat AID-expressing tumors. Given the growing list of tumor types with aberrant AID expression, this novel therapeutic approach has potential to impact a significant patient population.


Asunto(s)
Citidina Desaminasa/metabolismo , Recombinación Homóloga/genética , Leucemia Linfocítica Crónica de Células B/enzimología , Leucemia Linfocítica Crónica de Células B/genética , Ácido 4,4'-Diisotiocianostilbeno-2,2'-Disulfónico/farmacología , Transporte Activo de Núcleo Celular/efectos de los fármacos , Transporte Activo de Núcleo Celular/efectos de la radiación , Animales , Linfocitos B/efectos de los fármacos , Linfocitos B/enzimología , Linfocitos B/patología , Linfocitos B/efectos de la radiación , Muerte Celular/efectos de los fármacos , Muerte Celular/efectos de la radiación , Línea Celular Transformada , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Citidina Desaminasa/genética , Roturas del ADN de Doble Cadena/efectos de los fármacos , Roturas del ADN de Doble Cadena/efectos de la radiación , Reparación del ADN/efectos de los fármacos , Reparación del ADN/efectos de la radiación , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Regulación Leucémica de la Expresión Génica/efectos de la radiación , Histonas/metabolismo , Recombinación Homóloga/efectos de los fármacos , Recombinación Homóloga/efectos de la radiación , Humanos , Ratones , Recombinasa Rad51/metabolismo , Radiación Ionizante
16.
J Immunol ; 189(5): 2374-82, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22826323

RESUMEN

Activation-induced cytidine deaminase (AID) initiates DNA double-strand breaks (DSBs) in the IgH gene (Igh) to stimulate isotype class switch recombination (CSR), and widespread breaks in non-Igh (off-target) loci throughout the genome. Because the DSBs that initiate class switching occur during the G1 phase of the cell cycle, and are repaired via end joining, CSR is considered a predominantly G1 reaction. By contrast, AID-induced non-Igh DSBs are repaired by homologous recombination. Although little is known about the connection between the cell cycle and either induction or resolution of AID-mediated non-Igh DSBs, their repair by homologous recombination implicates post-G1 phases. Coordination of DNA breakage and repair during the cell cycle is critical to promote normal class switching and prevent genomic instability. To understand how AID-mediated events are regulated through the cell cycle, we have investigated G1-to-S control in AID-dependent genome-wide DSBs. We find that AID-mediated off-target DSBs, like those induced in the Igh locus, are generated during G1. These data suggest that AID-mediated DSBs can evade G1/S checkpoint activation and persist beyond G1, becoming resolved during S phase. Interestingly, DSB resolution during S phase can promote not only non-Igh break repair, but also Ig CSR. Our results reveal novel cell cycle dynamics in response to AID-initiated DSBs, and suggest that the regulation of the repair of these DSBs through the cell cycle may ensure proper class switching while preventing AID-induced genomic instability.


Asunto(s)
Citidina Desaminasa/fisiología , Roturas del ADN de Doble Cadena , Cambio de Clase de Inmunoglobulina/genética , Isotipos de Inmunoglobulinas/genética , Fase S/genética , Fase S/inmunología , Animales , Linfocitos B/citología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Células Cultivadas , Citidina Desaminasa/deficiencia , Citidina Desaminasa/genética , Reparación del ADN/genética , Reparación del ADN/inmunología , Fase G1/genética , Fase G1/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
17.
Nat Immunol ; 11(9): 820-6, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20657597

RESUMEN

Activation-induced cytidine deaminase (AID) is required for somatic hypermutation and immunoglobulin class switching in activated B cells. Because AID has no known target-site specificity, there have been efforts to identify non-immunoglobulin AID targets. We show here that AID acts promiscuously, generating widespread DNA double-strand breaks (DSBs), genomic instability and cytotoxicity in B cells with less homologous recombination ability. We demonstrate that the homologous-recombination factor XRCC2 suppressed AID-induced off-target DSBs, promoting B cell survival. Finally, we suggest that aberrations that affect human chromosome 7q36, including XRCC2, correlate with genomic instability in B cell cancers. Our findings demonstrate that AID has promiscuous genomic DSB-inducing activity, identify homologous recombination as a safeguard against off-target AID action, and have implications for genomic instability in B cell cancers.


Asunto(s)
Citidina Desaminasa/metabolismo , Roturas del ADN , Recombinación Genética/genética , Linfocitos B/inmunología , Ciclo Celular , Supervivencia Celular , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Citometría de Flujo , Inestabilidad Genómica , Humanos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
Mol Cell Biol ; 28(7): 2295-303, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18212067

RESUMEN

Primary immunodeficiencies are rare but serious diseases with diverse genetic causes. Accumulating evidence suggests that defects in DNA double-strand break (DSB) repair can underlie many of these syndromes. In this context, the nonhomologous end joining pathway of DSB repair is absolutely required for lymphoid development, but possible roles for the homologous recombination (HR) pathway have remained more controversial. While recent evidence suggests that HR may indeed be important to suppress lymphoid transformation, the specific relationship of HR to normal lymphocyte development remains unclear. We have investigated roles of the X-ray cross-complementing 2 (Xrcc2) HR gene in lymphocyte development. We show that HR is critical for normal B-cell development, with Xrcc2 nullizygosity leading to p53-dependent early S-phase arrest. In the absence of p53 (encoded by Trp53), Xrcc2-null B cells can fully develop but show high rates of chromosome and chromatid fragmentation. We present a molecular model wherein Xrcc2 is important to preserve or restore replication forks during rapid clonal expansion of developing lymphocytes. Our findings demonstrate a key role for HR in lymphoid development and suggest that Xrcc2 defects could underlie some human primary immunodeficiencies.


Asunto(s)
Linfocitos B/citología , Reparación del ADN , Proteínas de Unión al ADN/fisiología , Linfopoyesis/fisiología , Recombinación Genética , Animales , Células Cultivadas/citología , Aberraciones Cromosómicas , Rotura Cromosómica , Técnicas de Cocultivo , Eliminación de Gen , Genes p53 , Inmunoglobulina M/biosíntesis , Interleucina-7/metabolismo , Antígenos Comunes de Leucocito/biosíntesis , Hígado/citología , Hígado/embriología , Linfopoyesis/genética , Ratones , Ratones Noqueados , Células 3T3 NIH/metabolismo , Fase S , Homología de Secuencia de Ácido Nucleico , Proteína p53 Supresora de Tumor/fisiología
19.
Proc Natl Acad Sci U S A ; 104(42): 16627-32, 2007 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-17921248

RESUMEN

Lipodystrophies are syndromes of adipose tissue degeneration associated with severe defects in lipid and glucose homeostasis. We report here the generation and analysis of Pparg(ldi), a targeted allele that confers conditional dominant lipodystrophy in mice. The Pparg(ldi) allele was generated by insertion of the Tet activator (tTA) and a tTA-regulated Flag-Pparg1 transgene into the Pparg gene. Unexpectedly, tTA elicits mild lipodystrophy, insulin resistance, and dyslipidemia, and the Flag-PPARgamma1 transgene surprisingly exacerbates these traits. Doxycycline can both completely prevent and reverse these phenotypes, providing a mouse model of inducible lipodystrophy. Embryonic fibroblasts from either Pparg(ldi/+) or the phenotypically similar aP2-nSrebp1c (Sr) transgenic mice undergo robust adipogenesis, suggesting that neither strain develops lipodystrophy because of defective adipocyte differentiation. In addition, Pparg(ldi/+) adipose tissue shares extensive gene expression aberrations with that of Sr mice, authenticating the phenotype at the molecular level and revealing a common expression signature of lipodystrophic fat. Thus, the Pparg(ldi/+) mouse provides a conditional animal model for studying lipodystrophy and its associated physiology and gene expression.


Asunto(s)
Modelos Animales de Enfermedad , Lipodistrofia/genética , Ratones Transgénicos , PPAR gamma/genética , Adipogénesis/genética , Alelos , Animales , Doxiciclina/farmacología , Fibroblastos/metabolismo , Expresión Génica , Resistencia a la Insulina/genética , Lipodistrofia/patología , Ratones , Regiones Promotoras Genéticas/efectos de los fármacos , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Tetraciclina/farmacología , Transactivadores/genética
20.
Gene ; 405(1-2): 65-78, 2007 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-17949927

RESUMEN

HIV-1 transcription is essential for the virus replication cycle. HIV-1 Tat is a viral transactivator that strongly stimulates the processivity of RNA polymerase II (RNAPII) via recruitment of the cyclin T1/CDK9 positive transcription elongation factor, which phosphorylates the C-terminal domain (CTD) of RNAPII. Consistently, HIV-1 replication in transformed cells is very sensitive to direct CDK9 inhibition. Thus, CDK9 could be a potential target for anti-HIV-1 therapy. A clearer understanding of the requirements for CDK9 activity in primary human T cells is needed to assess whether the CDK9-dependent step in HIV-1 transcription can be targeted clinically. We have investigated the effects of limiting CDK9 activity with recombinant lentiviruses expressing a dominant-negative form of CDK9 (HA-dnCDK9) in peripheral blood lymphocytes (PBLs) and other cells. Our results show that direct inhibition of CDK9 potently inhibits HIV-1 replication in single-round infection assays with little to undetectable effects on RNAPII transcription, RNA synthesis, proliferation and viability. In PBLs purified from multiple donors, direct inhibition of CDK9 activity blocks HIV-1 replication/transcription but does not prevent T-cell activation, as determined via measurement of cell surface and cell cycle entry and progression markers, and DNA synthesis. We have also compared the effects of HA-dnCDK9 to flavopiridol (FVP), a general CDK inhibitor that potently inhibits CDK9. In contrast to HA-dnCDK9, FVP interferes with key cellular processes at concentrations that inhibit HIV-1 replication with potency similar to HA-dnCDK9. In particular, FVP inhibits several T-cell activation markers and DNA synthesis in primary PBLs at the minimal concentrations required to inhibit HIV-1 replication. Our results imply that small pharmacological compounds targeting CDK9 with enhanced selectivity could be developed into effective anti-HIV-1 therapeutic drugs.


Asunto(s)
Quinasa 9 Dependiente de la Ciclina/antagonistas & inhibidores , VIH-1/fisiología , Activación de Linfocitos/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Linfocitos T/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Línea Celular , Citometría de Flujo , VIH-1/genética , Humanos , Linfocitos T/enzimología , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...