Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 588(7837): 303-307, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33239790

RESUMEN

The gut microbiota influences development1-3 and homeostasis4-7 of the mammalian immune system, and is associated with human inflammatory8 and immune diseases9,10 as well as responses to immunotherapy11-14. Nevertheless, our understanding of how gut bacteria modulate the immune system remains limited, particularly in humans, where the difficulty of direct experimentation makes inference challenging. Here we study hundreds of hospitalized-and closely monitored-patients with cancer receiving haematopoietic cell transplantation as they recover from chemotherapy and stem-cell engraftment. This aggressive treatment causes large shifts in both circulatory immune cell and microbiota populations, enabling the relationships between the two to be studied simultaneously. Analysis of observed daily changes in circulating neutrophil, lymphocyte and monocyte counts and more than 10,000 longitudinal microbiota samples revealed consistent associations between gut bacteria and immune cell dynamics. High-resolution clinical metadata and Bayesian inference allowed us to compare the effects of bacterial genera in relation to those of immunomodulatory medications, revealing a considerable influence of the gut microbiota-together and over time-on systemic immune cell dynamics. Our analysis establishes and quantifies the link between the gut microbiota and the human immune system, with implications for microbiota-driven modulation of immunity.


Asunto(s)
Microbioma Gastrointestinal/inmunología , Leucocitos/citología , Leucocitos/inmunología , Factores de Edad , Teorema de Bayes , Trasplante de Microbiota Fecal , Femenino , Humanos , Recuento de Leucocitos , Linfocitos/citología , Linfocitos/inmunología , Monocitos/citología , Monocitos/inmunología , Neutrófilos/citología , Neutrófilos/inmunología , Reproducibilidad de los Resultados
2.
Genes Chromosomes Cancer ; 63(7): e23257, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39031442

RESUMEN

Gene panel sequencing has become a common diagnostic tool for detecting somatically acquired mutations in myeloid neoplasms. However, many panels have restricted content, provide insufficient sensitivity levels, or lack clinically validated workflows. We here describe the development and validation of the Genomic Medicine Sweden myeloid gene panel (GMS-MGP), a capture-based 191 gene panel including mandatory genes in contemporary guidelines as well as emerging candidates. The GMS-MGP displayed uniform coverage across all targets, including recognized difficult GC-rich areas. The validation of 117 previously described somatic variants showed a 100% concordance with a limit-of-detection of a 0.5% variant allele frequency (VAF), achieved by utilizing error correction and filtering against a panel-of-normals. A national interlaboratory comparison investigating 56 somatic variants demonstrated highly concordant results in both detection rate and reported VAFs. In addition, prospective analysis of 323 patients analyzed with the GMS-MGP as part of standard-of-care identified clinically significant genes as well as recurrent mutations in less well-studied genes. In conclusion, the GMS-MGP workflow supports sensitive detection of all clinically relevant genes, facilitates novel findings, and is, based on the capture-based design, easy to update once new guidelines become available. The GMS-MGP provides an important step toward nationally harmonized precision diagnostics of myeloid malignancies.


Asunto(s)
Medicina de Precisión , Humanos , Medicina de Precisión/métodos , Mutación , Suecia , Pruebas Genéticas/métodos , Pruebas Genéticas/normas , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/diagnóstico , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Frecuencia de los Genes
3.
Blood ; 139(18): 2758-2769, 2022 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-35061893

RESUMEN

Low intestinal microbial diversity is associated with poor outcomes after allogeneic hematopoietic cell transplantation (HCT). Using 16S rRNA sequencing of 2067 stool samples and flow cytometry data from 2370 peripheral blood samples drawn from 894 patients who underwent allogeneic HCT, we have linked features of the early post-HCT microbiome with subsequent immune cell recovery. We examined lymphocyte recovery and microbiota features in recipients of both unmodified and CD34-selected allografts. We observed that fecal microbial diversity was an independent predictor of CD4 T-cell count 3 months after HCT in recipients of a CD34-selected allograft, who are dependent on de novo lymphopoiesis for their immune recovery. In multivariate models using clinical factors and microbiota features, we consistently observed that increased fecal relative abundance of genus Staphylococcus during the early posttransplant period was associated with worse CD4 T-cell recovery. Our observations suggest that the intestinal bacteria, or the factors they produce, can affect early lymphopoiesis and the homeostasis of allograft-derived T cells after transplantation.


Asunto(s)
Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Linfocitos T CD4-Positivos , Humanos , Recuento de Linfocitos , ARN Ribosómico 16S , Trasplante Homólogo
4.
Br J Haematol ; 201(3): 480-488, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36650117

RESUMEN

The tick-borne pathogen Neoehrlichia (N.) mikurensis is implicated in persistent infection of the vascular endothelium. B cells are crucial for the host defence to this infection. Chronic stimulation of B cells may result in B-cell transformation and lymphoma. Five patients with malignant B-cell lymphoma and concomitant N. mikurensis infection were investigated regarding clinical picture, lymphoma subtype, B-cell lymphoma immunophenotype and IGHV (variable region of the immunoglobulin heavy) gene repertoire. Three of the five patients improved markedly and ceased lymphoma treatment after doxycycline treatment to eliminate N. mikurensis. Sequencing the B-cell lymphoma IGHV genes revealed preferred usage of the IGHV1 (IGHV1-2, and -69) and IGHV3 (IGHV3-15, -21, -23) families. In conclusion, N. mikurensis infection may drive the development of malignant B-cell lymphomas. Eradication of the pathogen appears to induce remission with apparent curing of the lymphoma in some cases.


Asunto(s)
Infecciones por Anaplasmataceae , Linfoma de Células B , Enfermedades por Picaduras de Garrapatas , Infecciones por Anaplasmataceae/complicaciones , Infecciones por Anaplasmataceae/tratamiento farmacológico , Infecciones por Anaplasmataceae/microbiología , Linfoma de Células B/etiología , Linfoma de Células B/genética , Enfermedades por Picaduras de Garrapatas/microbiología , Receptores de Antígenos de Linfocitos B , Doxiciclina/uso terapéutico , Antibacterianos/uso terapéutico , Humanos , Masculino , Femenino , Persona de Mediana Edad , Anciano , Análisis de Secuencia de ADN , Inmunofenotipificación
5.
Int J Cancer ; 151(5): 770-782, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35583991

RESUMEN

Acute myeloid leukemia (AML) results from aberrant hematopoietic processes and these changes are frequently initiated by chromosomal translocations. One particular subtype, AML with translocation t(7;12)(q36;p13), is found in children diagnosed before 2 years of age. The mechanisms for leukemogenesis induced by t(7;12) is not understood, in part because of the lack of efficient methods to reconstruct the leukemia-associated genetic aberration with correct genomic architecture and regulatory elements. We therefore created induced pluripotent stem cell (iPSC) lines that carry the translocation t(7;12) using CRISPR/Cas9. These t(7;12) iPSC showed propensity to differentiate into all three germ layers, confirming retained stem cell properties. The potential for differentiation into hematopoietic stem and progenitor cells (HSPC) was shown by expression of CD34, CD43 and CD45. Compared with the parental iPSC line, a significant decrease in cells expressing CD235a and CD41a was seen in the t(7;12) iPSC-derived HSPC (iHSPC), suggesting a block in differentiation. Moreover, colony formation assay showed an accumulation of cells at the erythroid and myeloid progenitor stages. Gene expression analysis revealed significant down-regulation of genes associated with megakaryocyte differentiation and up-regulation of genes associated with myeloid pathways but also genes typically seen in AML cases with t(7;12). Thus, this iPSC t(7;12) leukemia model of the t(7;12) AML subtype constitutes a valuable tool for further studies of the mechanisms for leukemia development and to find new treatment options.


Asunto(s)
Diferenciación Celular , Proteínas de Homeodominio , Células Madre Pluripotentes Inducidas , Leucemia Mieloide Aguda , Células Progenitoras de Megacariocitos y Eritrocitos , Factores de Transcripción , Diferenciación Celular/genética , Niño , Expresión Génica/genética , Expresión Génica/fisiología , Perfilación de la Expresión Génica , Células Madre Hematopoyéticas/fisiología , Proteínas de Homeodominio/genética , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Leucemia Mieloide Aguda/genética , Células Progenitoras de Megacariocitos y Eritrocitos/fisiología , Megacariocitos/fisiología , Factores de Transcripción/genética , Translocación Genética
6.
Blood ; 129(8): 927-933, 2017 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-27940475

RESUMEN

Hematopoietic cell transplantation (HCT) is a critical treatment of patients with high-risk hematopoietic malignancies, hematological deficiencies, and other immune diseases. In allogeneic HCT (allo-HCT), donor-derived T cells recognize host tissues as foreign, causing graft-versus-host disease (GVHD) which is a main contributor to morbidity and mortality. The intestine is one of the organs most severely affected by GVHD and research has recently highlighted the importance of bacteria, particularly the gut microbiota, in HCT outcome and in GVHD development. Loss of intestinal bacterial diversity is common during the course of HCT and is associated with GVHD development and treatment with broad-spectrum antibiotics. Loss of intestinal diversity and outgrowth of opportunistic pathogens belonging to the phylum Proteobacteria and Enterococcus genus have also been linked to increased treatment-related mortality including GVHD, infections, and organ failure after allo-HCT. Experimental studies in allo-HCT animal models have shown some promising results for prebiotic and probiotic strategies as prophylaxis or treatment of GVHD. Continuous research will be important to define the relation of cause and effect for these associations between microbiota features and HCT outcomes. Importantly, studies focused on geographic and cultural differences in intestinal microbiota are necessary to define applicability of new strategies targeting the intestinal microbiota.


Asunto(s)
Microbioma Gastrointestinal , Enfermedad Injerto contra Huésped/microbiología , Trasplante de Células Madre Hematopoyéticas , Intestinos/microbiología , Animales , Enfermedad Injerto contra Huésped/complicaciones , Enfermedad Injerto contra Huésped/fisiopatología , Enfermedad Injerto contra Huésped/terapia , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Intestinos/fisiopatología , Estado Nutricional , Acondicionamiento Pretrasplante/métodos , Trasplante Homólogo
7.
Haematologica ; 103(2): 246-255, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29217774

RESUMEN

Micro-ribonucleic acid-155 (miR-155) is one of the first described oncogenic miRNAs. Although multiple direct targets of miR-155 have been identified, it is not clear how it contributes to the pathogenesis of acute myeloid leukemia. We found miR-155 to be a direct target of Meis1 in murine Hoxa9/Meis1 induced acute myeloid leukemia. The additional overexpression of miR-155 accelerated the formation of acute myeloid leukemia in Hoxa9 as well as in Hoxa9/Meis1 cells in vivo However, in the absence or following the removal of miR-155, leukemia onset and progression were unaffected. Although miR-155 accelerated growth and homing in addition to impairing differentiation, our data underscore the pathophysiological relevance of miR-155 as an accelerator rather than a driver of leukemogenesis. This further highlights the complexity of the oncogenic program of Meis1 to compensate for the loss of a potent oncogene such as miR-155. These findings are highly relevant to current and developing approaches for targeting miR-155 in acute myeloid leukemia.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Leucemia Mieloide Aguda/etiología , MicroARNs/antagonistas & inhibidores , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/farmacología , Animales , Carcinogénesis/genética , Regulación Leucémica de la Expresión Génica , Humanos , Leucemia Mieloide Aguda/genética , Ratones , MicroARNs/metabolismo
8.
Circ Res ; 115(9): 781-9, 2014 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-25212213

RESUMEN

RATIONALE: Cell proliferation and cell cycle control mechanisms are thought to play central roles in the pathogenesis of atherosclerosis. The transcription factor Zinc finger protein 148 (Zfp148) was shown recently to maintain cell proliferation under oxidative conditions by suppressing p53, a checkpoint protein that arrests proliferation in response to various stressors. It is established that inactivation of p53 accelerates atherosclerosis, but whether increased p53 activation confers protection against the disease remains to be determined. OBJECTIVE: We aimed to test the hypothesis that Zfp148 deficiency reduces atherosclerosis by unleashing p53 activity. METHODS AND RESULTS: Mice harboring a gene-trap mutation in the Zfp148 locus (Zfp148(gt/+)) were bred onto the apolipoprotein E (Apoe)(-/-) genetic background and fed a high-fat or chow diet. Loss of 1 copy of Zfp148 markedly reduced atherosclerosis without affecting lipid metabolism. Bone marrow transplantation experiments revealed that the effector cell is of hematopoietic origin. Peritoneal macrophages and atherosclerotic lesions from Zfp148(gt/+)Apoe(-/-) mice showed increased levels of phosphorylated p53 compared with controls, and atherosclerotic lesions contained fewer proliferating macrophages. Zfp148(gt/+)Apoe(-/-) mice were further crossed with p53-null mice (Trp53(-/-) [the gene encoding p53]). There was no difference in atherosclerosis between Zfp148(gt/+)Apoe(-/-) mice and controls on a Trp53(+/-) genetic background, and there was no difference in levels of phosphorylated p53 or cell proliferation. CONCLUSIONS: Zfp148 deficiency increases p53 activity and protects against atherosclerosis by causing proliferation arrest of lesional macrophages, suggesting that drugs targeting macrophage proliferation may be useful in the treatment of atherosclerosis.


Asunto(s)
Enfermedades de la Aorta/prevención & control , Aterosclerosis/prevención & control , Puntos de Control del Ciclo Celular , Proliferación Celular , Proteínas de Unión al ADN/deficiencia , Macrófagos Peritoneales/metabolismo , Factores de Transcripción/deficiencia , Activación Transcripcional , Proteína p53 Supresora de Tumor/metabolismo , Animales , Enfermedades de la Aorta/etiología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/patología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/etiología , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Trasplante de Médula Ósea , Enfermedades de las Arterias Carótidas/metabolismo , Enfermedades de las Arterias Carótidas/patología , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Humanos , Macrófagos Peritoneales/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Placa Aterosclerótica , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/genética
9.
Pediatr Blood Cancer ; 61(3): 424-30, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24424791

RESUMEN

BACKGROUND: In children, T-cell acute lymphoblastic leukemia (T-ALL) has inferior prognosis compared with B-cell precursor ALL. In order to improve survival, individualized treatment strategies and thus risk stratification algorithms are warranted, ideally already at the time of diagnosis. PROCEDURE: We analyzed the frequency and prognostic implication of mutations in NOTCH1 and FBXW7 in 79 cases of Swedish childhood T-ALL treated according to the Nordic Society of Pediatric Hematology and Oncology (NOPHO) ALL-1992 and ALL-2000 protocols. In a subgroup of patients, we also investigated the functional relevance of NOTCH1 mutations measured as expression of the HES1, MYB, and MYC genes. RESULTS: Forty-seven of the cases (59%) displayed mutations in NOTCH1 and/or FBXW7. There was no difference in overall (P = 0.14) or event-free survival (EFS) (P = 0.10) in patients with T-ALL with mutation(s) in NOTCH1/FBXW7 compared with patients with T-ALL without mutations in any of these genes. T-ALL carrying NOTCH1 mutations had increased HES1 and MYB mRNA expression (HES1 9.2 ± 1.9 (mean ± SEM), MYB 8.7 ± 0.8 (mean ± SEM)) compared to T-ALL with wild-type NOTCH1 (HES1 1.8 ± 0.7, MYB 5.1 ± 1.2, P = 0.02 and 0.008, respectively). In cases of T-ALL with high HES1 expression, improved overall (P = 0.02) and EFS (P = 0.028) was seen. CONCLUSIONS: Increased NOTCH activity, reflected by increased HES1 expression, is associated with improved outcome in pediatric T-ALL, but its role as a diagnostic tool or a therapeutic target in future clinical treatment protocols remains to be elucidated.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proteínas F-Box/genética , Mutación , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Receptor Notch1/genética , Ubiquitina-Proteína Ligasas/genética , Adolescente , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Niño , Preescolar , Proteína 7 que Contiene Repeticiones F-Box-WD , Femenino , Genes myb , Proteínas de Homeodominio/genética , Humanos , Lactante , Masculino , Leucemia-Linfoma Linfoblástico de Células T Precursoras/mortalidad , Leucemia-Linfoma Linfoblástico de Células T Precursoras/terapia , Pronóstico , Factor de Transcripción HES-1
10.
Blood ; 118(22): 5905-13, 2011 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-21967978

RESUMEN

Mutation status of FLT3, NPM1, CEBPA, and WT1 genes and gene expression levels of ERG, MN1, BAALC, FLT3, and WT1 have been identified as possible prognostic markers in acute myeloid leukemia (AML). We have performed a thorough prognostic evaluation of these genetic markers in patients with pediatric AML enrolled in the Nordic Society of Pediatric Hematology and Oncology (NOPHO) 1993 or NOPHO 2004 protocols. Mutation status and expression levels were analyzed in 185 and 149 patients, respectively. Presence of FLT3-internal tandem duplication (ITD) was associated with significantly inferior event-free survival (EFS), whereas presence of an NPM1 mutation in the absence of FLT3-ITD correlated with significantly improved EFS. Furthermore, high levels of ERG and BAALC transcripts were associated with inferior EFS. No significant correlation with survival was seen for mutations in CEBPA and WT1 or with gene expression levels of MN1, FLT3, and WT1. In multivariate analysis, the presence of FLT3-ITD and high BAALC expression were identified as independent prognostic markers of inferior EFS. We conclude that analysis of the mutational status of FLT3 and NPM1 at diagnosis is important for prognostic stratification of patients with pediatric AML and that determination of the BAALC gene expression level can add valuable information.


Asunto(s)
Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/genética , Proteínas de Neoplasias/genética , Secuencias Repetidas en Tándem/genética , Tirosina Quinasa 3 Similar a fms/genética , Adolescente , Edad de Inicio , Biomarcadores de Tumor/genética , Niño , Preescolar , Regulación Leucémica de la Expresión Génica , Humanos , Lactante , Recién Nacido , Leucemia Mieloide Aguda/epidemiología , Mutación , Nucleofosmina , Pronóstico , Regulación hacia Arriba/genética
11.
Front Oncol ; 13: 1217712, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37664045

RESUMEN

Introduction: The suitability of whole-genome sequencing (WGS) as the sole method to detect clinically relevant genomic aberrations in B-cell acute lymphoblastic leukemia (ALL) was investigated with the aim of replacing current diagnostic methods. Methods: For this purpose, we assessed the analytical performance of 150 bp paired-end WGS (90x leukemia/30x germline). A set of 88 retrospective B-cell ALL samples were selected to represent established ALL subgroups as well as ALL lacking stratifying markers by standard-of-care (SoC), so-called B-other ALL. Results: Both the analysis of paired leukemia/germline (L/N)(n=64) as well as leukemia-only (L-only)(n=88) detected all types of aberrations mandatory in the current ALLTogether trial protocol, i.e., aneuploidies, structural variants, and focal copy-number aberrations. Moreover, comparison to SoC revealed 100% concordance and that all patients had been assigned to the correct genetic subgroup using both approaches. Notably, WGS could allocate 35 out of 39 B-other ALL samples to one of the emerging genetic subgroups considered in the most recent classifications of ALL. We further investigated the impact of high (90x; n=58) vs low (30x; n=30) coverage on the diagnostic yield and observed an equally perfect concordance with SoC; low coverage detected all relevant lesions. Discussion: The filtration of the WGS findings with a short list of genes recurrently rearranged in ALL was instrumental to extract the clinically relevant information efficiently. Nonetheless, the detection of DUX4 rearrangements required an additional customized analysis, due to multiple copies of this gene embedded in the highly repetitive D4Z4 region. We conclude that the diagnostic performance of WGS as the standalone method was remarkable and allowed detection of all clinically relevant genomic events in the diagnostic setting of B-cell ALL.

12.
Genes (Basel) ; 12(6)2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-34200630

RESUMEN

Pediatric acute myeloid leukemia (AML) is a heterogeneous disease composed of clinically relevant subtypes defined by recurrent cytogenetic aberrations. The majority of the aberrations used in risk grouping for treatment decisions are extensively studied, but still a large proportion of pediatric AML patients remain cytogenetically undefined and would therefore benefit from additional molecular investigation. As aberrant epigenetic regulation has been widely observed during leukemogenesis, we hypothesized that DNA methylation signatures could be used to predict molecular subtypes and identify signatures with prognostic impact in AML. To study genome-wide DNA methylation, we analyzed 123 diagnostic and 19 relapse AML samples on Illumina 450k DNA methylation arrays. We designed and validated DNA methylation-based classifiers for AML cytogenetic subtype, resulting in an overall test accuracy of 91%. Furthermore, we identified methylation signatures associated with outcome in t(8;21)/RUNX1-RUNX1T1, normal karyotype, and MLL/KMT2A-rearranged subgroups (p < 0.01). Overall, these results further underscore the clinical value of DNA methylation analysis in AML.


Asunto(s)
Biomarcadores de Tumor/genética , Metilación de ADN , Epigenoma , Leucemia Mieloide Aguda/genética , Adolescente , Niño , Preescolar , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Femenino , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Lactante , Leucemia Mieloide Aguda/clasificación , Leucemia Mieloide Aguda/patología , Masculino , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas de Fusión Oncogénica/genética , Proteína 1 Compañera de Translocación de RUNX1/genética
13.
Semin Hematol ; 57(1): 7-12, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-32690142

RESUMEN

Clonal hematopoiesis of indetermined potential (CHIP) is increasingly common with age and identified in more than 1 in 10 healthy individuals at the age of 70. Mutations in epigenetic and splicing factors are recurrent genetic events in CHIP, and experimental data suggest that microbial and inflammatory factors may contribute to the selective expansion of hematopoietic stem cells carrying these mutations. In parallel, CHIP is associated with an increased incidence of cardiovascular disease and studies in mice support a causal relationship where mutated hematopoietic cells contribute to inflammation and atherosclerotic plaque formation. Collectively, current clinical and experimental data suggest a complex network where genetic alterations and inflammatory factors contribute to the development of the early stages of hematological malignancy.


Asunto(s)
Hematopoyesis Clonal/genética , Inflamación/genética , Leucemia/genética , Humanos , Mutación
14.
Sci Rep ; 10(1): 14156, 2020 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-32843651

RESUMEN

Recent data suggest that the transcription factor Zfp148 represses activation of the tumor suppressor p53 in mice and that therapeutic targeting of the human orthologue ZNF148 could activate the p53 pathway without causing detrimental side effects. We have previously shown that Zfp148 deficiency promotes p53-dependent proliferation arrest of mouse embryonic fibroblasts (MEFs), but the underlying mechanism is not clear. Here, we showed that Zfp148 deficiency downregulated cell cycle genes in MEFs in a p53-dependent manner. Proliferation arrest of Zfp148-deficient cells required increased expression of ARF, a potent activator of the p53 pathway. Chromatin immunoprecipitation showed that Zfp148 bound to the ARF promoter, suggesting that Zfp148 represses ARF transcription. However, Zfp148 preferentially bound to promoters of other transcription factors, indicating that deletion of Zfp148 may have pleiotropic effects that activate ARF and p53 indirectly. In line with this, we found no evidence of genetic interaction between TP53 and ZNF148 in CRISPR and siRNA screen data from hundreds of human cancer cell lines. We conclude that Zfp148 deficiency, by increasing ARF transcription, downregulates cell cycle genes and cell proliferation in a p53-dependent manner. However, the lack of genetic interaction between ZNF148 and TP53 in human cancer cells suggests that therapeutic targeting of ZNF148 may not increase p53 activity in humans.


Asunto(s)
Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica/genética , Transducción de Señal/genética , Factores de Transcripción/genética , Proteína p53 Supresora de Tumor/fisiología , Animales , Sistemas CRISPR-Cas , Puntos de Control del Ciclo Celular/genética , Proteínas de Ciclo Celular/biosíntesis , Proteínas de Ciclo Celular/genética , División Celular , Línea Celular , Inmunoprecipitación de Cromatina , Cisplatino/toxicidad , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Daño del ADN , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/fisiología , Regulación hacia Abajo , Factores de Transcripción E2F/fisiología , Etopósido/toxicidad , Fibroblastos , Ontología de Genes , Ratones , Interferencia de ARN , ARN Interferente Pequeño/genética , Factores de Transcripción/deficiencia , Factores de Transcripción/fisiología
15.
Leukemia ; 34(5): 1253-1265, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31768018

RESUMEN

MicroRNAs (miRNAs) are commonly deregulated in acute myeloid leukemia (AML), affecting critical genes not only through direct targeting, but also through modulation of downstream effectors. Homeobox (Hox) genes balance self-renewal, proliferation, cell death, and differentiation in many tissues and aberrant Hox gene expression can create a predisposition to leukemogenesis in hematopoietic cells. However, possible linkages between the regulatory pathways of Hox genes and miRNAs are not yet fully resolved. We identified miR-708 to be upregulated in Hoxa9/Meis1 AML inducing cell lines as well as in AML patients. We further showed Meis1 directly targeting miR-708 and modulating its expression through epigenetic transcriptional regulation. CRISPR/Cas9 mediated knockout of miR-708 in Hoxa9/Meis1 cells delayed disease onset in vivo, demonstrating for the first time a pro-leukemic contribution of miR-708 in this context. Overexpression of miR-708 however strongly impeded Hoxa9 mediated transformation and homing capacity in vivo through modulation of adhesion factors and induction of myeloid differentiation. Taken together, we reveal miR-708, a putative tumor suppressor miRNA and direct target of Meis1, as a potent antagonist of the Hoxa9 phenotype but an effector of transformation in Hoxa9/Meis1. This unexpected finding highlights the yet unexplored role of miRNAs as indirect regulators of the Hox program during normal and aberrant hematopoiesis.


Asunto(s)
Regulación Leucémica de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Leucemia Mieloide Aguda/patología , MicroARNs/genética , Células Mieloides/patología , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/metabolismo , Animales , Apoptosis , Sistemas CRISPR-Cas , Diferenciación Celular , Proliferación Celular , Femenino , Hematopoyesis , Proteínas de Homeodominio/genética , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Ratones , Ratones Endogámicos C57BL , MicroARNs/antagonistas & inhibidores , MicroARNs/metabolismo , Células Mieloides/metabolismo , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/genética , Células Tumorales Cultivadas
16.
Bone Marrow Transplant ; 54(Suppl 2): 756-758, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31431719

RESUMEN

Recent studies in both mice and humans have demonstrated that the intestinal microbiota can affect hematopoiesis. Here, we performed experiments in preclinical mouse models for syngeneic and allogeneic HCT. To study the metabolic effects of intestinal flora depletion on post-transplant hematopoiesis in humans, we performed HCT experiments using a metabolic chamber and bomb calorimetry of feces. Taken together, we show that the intestinal microbiota supports post-transplant hematopoietic reconstitution in HCT recipients through its role in dietary energy uptake.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Hematopoyesis/fisiología , Trasplante de Células Madre Hematopoyéticas/métodos , Acondicionamiento Pretrasplante/métodos , Animales , Humanos , Ratones
17.
Oncogene ; 38(9): 1534-1543, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30323311

RESUMEN

Mutations leading to constitutive RAS activation contribute in myeloid leukemogenesis. RAS mutations in myeloid cells are accompanied by excessive formation of reactive oxygen species (ROS), but the source of ROS and their role for the initiation and progression of leukemia have not been clearly defined. To determine the role of NOX2-derived ROS in RAS-driven leukemia, double transgenic LSL-KrasG12D × Mx1-Cre mice expressing oncogenic KRAS in hematopoietic cells (M-KrasG12D) were treated with Nα-methyl-histamine (NMH) that targeted the production of NOX2-derived ROS in leukemic cells by agonist activity at histamine H2 receptors. M-KrasG12D mice developed myeloid leukemia comprising mature CD11b+Gr1+ myeloid cells that produced NOX2-derived ROS. Treatment of M-KrasG12D mice with NMH delayed the development of myeloproliferative disease and prolonged survival. In addition, NMH-treated M-KrasG12D mice showed reduction of intracellular ROS along with reduced DNA oxidation and reduced occurence of double-stranded DNA breaks in myeloid cells. The in vivo expansion of leukemia was markedly reduced in triple transgenic mice where KRAS was expressed in hematopoietic cells of animals with genetic NOX2 deficiency (Nox2-/- × LSL-KrasG12D × Mx1-Cre). Treatment with NMH did not alter in vivo expansion of leukemia in these NOX2-deficient transgenic mice. We propose that NOX2-derived ROS may contribute to the progression of KRAS-induced leukemia and that strategies to target NOX2 merit further evaluation in RAS-mutated hematopoietic cancer.


Asunto(s)
Hematopoyesis/genética , Trastornos Mieloproliferativos/genética , NADPH Oxidasa 2/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Animales , Proliferación Celular , Transformación Celular Neoplásica/genética , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos , Mutación , Trastornos Mieloproliferativos/patología , Estrés Oxidativo/genética , Especies Reactivas de Oxígeno/metabolismo , Análisis de Supervivencia
18.
Leuk Res ; 75: 61-68, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30384975

RESUMEN

Endothelin receptor type A (EDNRA) is known as a mediator of cell proliferation and survival. Aberrant regulation of EDNRA has been shown to play a role in tumor growth and metastasis. Using a global gene expression screen, we found that expression of Ednra was upregulated in murine leukemia inducing cells co-expressing Hoxa9 and Meis1 compared to cells only expressing Hoxa9. The aim of this study was to explore the role of Ednra in leukemogenesis further. In a murine bone marrow transplantation model, mice transplanted with cells overexpressing Ednra and Hoxa9 succumbed to leukemia significantly earlier than mice transplanted with cells overexpressing Hoxa9 only. Furthermore, overexpression of Ednra led to increased proliferation and resistance to apoptosis of bone marrow cells in vitro. We could also show that Meis1 binds to the Ednra promoter region, suggesting a regulatory role for Meis1 in Ednra expression. Taken together, our results suggest a role for Ednra in Hoxa9/Meis1-driven leukemogenesis.


Asunto(s)
Regulación Leucémica de la Expresión Génica/fisiología , Proteínas de Homeodominio/metabolismo , Leucemia Mieloide Aguda/metabolismo , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/metabolismo , Receptor de Endotelina A/metabolismo , Animales , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos C57BL
19.
Cell Host Microbe ; 23(4): 447-457.e4, 2018 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-29576480

RESUMEN

Bone marrow transplantation (BMT) offers curative potential for patients with high-risk hematologic malignancies, but the post-transplantation period is characterized by profound immunodeficiency. Recent studies indicate that the intestinal microbiota not only regulates mucosal immunity, but can also contribute to systemic immunity and hematopoiesis. Using antibiotic-mediated microbiota depletion in a syngeneic BMT mouse model, here we describe a role for the intestinal flora in hematopoietic recovery after BMT. Depletion of the intestinal microbiota resulted in impaired recovery of lymphocyte and neutrophil counts, while recovery of the hematopoietic stem and progenitor compartments and the erythroid lineage were largely unaffected. Depletion of the intestinal microbiota also reduced dietary energy uptake and visceral fat stores. Caloric supplementation through sucrose in the drinking water improved post-BMT hematopoietic recovery in mice with a depleted intestinal flora. Taken together, we show that the intestinal microbiota contribute to post-BMT hematopoietic reconstitution in mice through improved dietary energy uptake.


Asunto(s)
Trasplante de Médula Ósea , Microbioma Gastrointestinal , Apoyo Nutricional , Animales , Médula Ósea/fisiología , Hematopoyesis , Ratones , Modelos Animales , Resultado del Tratamiento
20.
J Clin Oncol ; 35(15): 1650-1659, 2017 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-28296584

RESUMEN

Purpose The major causes of mortality after allogeneic hematopoietic-cell transplantation (allo-HCT) are relapse, graft-versus-host disease (GVHD), and infection. We have reported previously that alterations in the intestinal flora are associated with GVHD, bacteremia, and reduced overall survival after allo-HCT. Because intestinal bacteria are potent modulators of systemic immune responses, including antitumor effects, we hypothesized that components of the intestinal flora could be associated with relapse after allo-HCT. Methods The intestinal microbiota of 541 patients admitted for allo-HCT was profiled by means of 16S ribosomal sequencing of prospectively collected stool samples. We examined the relationship between abundance of microbiota species or groups of related species and relapse/progression of disease during 2 years of follow-up time after allo-HCT by using cause-specific proportional hazards in a retrospective discovery-validation cohort study. Results Higher abundance of a bacterial group composed mostly of Eubacterium limosum in the validation set was associated with a decreased risk of relapse/progression of disease (hazard ratio [HR], 0.82 per 10-fold increase in abundance; 95% CI, 0.71 to 0.95; P = .009). When the patients were categorized according to presence or absence of this bacterial group, presence also was associated with less relapse/progression of disease (HR, 0.52; 95% CI, 0.31 to 0.87; P = .01). The 2-year cumulative incidences of relapse/progression among patients with and without this group of bacteria were 19.8% and 33.8%, respectively. These associations remained significant in multivariable models and were strongest among recipients of T-cell-replete allografts. Conclusion We found associations between the abundance of a group of bacteria in the intestinal flora and relapse/progression of disease after allo-HCT. These might serve as potential biomarkers or therapeutic targets to prevent relapse and improve survival after allo-HCT.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Neoplasias/microbiología , Neoplasias/cirugía , Biomarcadores de Tumor/metabolismo , Heces/microbiología , Femenino , Enfermedad Injerto contra Huésped/microbiología , Humanos , Masculino , Persona de Mediana Edad , Neoplasias/metabolismo , Estudios Retrospectivos , Trasplante Homólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA