Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 10(1): 2851, 2020 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-32071334

RESUMEN

Epigenetic reprogramming in Acute Myeloid Leukemia (AML) leads to the aberrant activation of super enhancer (SE) landscapes that drive the expression of key oncogenes, including the oncogenic MYC pathway. These SEs have been identified as promising therapeutic targets, and have given rise to a new class of drugs, including BET protein inhibitors, which center on targeting SE activity. NR4A nuclear receptors are tumor suppressors of AML that function in part through transcriptional repression of the MYC-driven oncogenic program via mechanisms that remain unclear. Here we show that NR4A1, and the NR4A inducing drug dihydroergotamine (DHE), regulate overlapping gene expression programs in AML and repress transcription of a subset of SE-associated leukemic oncogenes, including MYC. NR4As interact with an AML-selective SE cluster that governs MYC transcription and decommissions its activation status by dismissing essential SE-bound coactivators including BRD4, Mediator and p300, leading to loss of p300-dependent H3K27 acetylation and Pol 2-dependent eRNA transcription. DHE shows similar efficacy to the BET inhibitor JQ1 at repressing SE-dependent MYC expression and AML growth in mouse xenografts. Thus, DHE induction of NR4As provides an alternative strategy to BET inhibitors to target MYC dependencies via suppression of the AML-selective SE governing MYC expression.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Leucemia Mieloide Aguda/tratamiento farmacológico , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Proteínas Proto-Oncogénicas c-myc/genética , Animales , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Reprogramación Celular/genética , Dihidroergotamina/farmacología , Proteína p300 Asociada a E1A/genética , Epigénesis Genética/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Ratones , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/agonistas , Oncogenes/genética , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/genética , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Leukemia ; 33(1): 52-63, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29884904

RESUMEN

NR4As are AML tumor suppressors that are frequently silenced in human acute myeloid leukemia (AML). Despite their potential as novel targets for therapeutic intervention, mechanisms of NR4A silencing and strategies for their reactivation remain poorly defined. Here we show that NR4A silencing in AML occurs through blockade of transcriptional elongation rather than epigenetic promoter silencing. By intersection of NR4A-regulated gene signatures captured upon acute, exogenous expression of NR4As in human AML cells with in silico chemical genomics screening, we identify several FDA-approved drugs including dihydroergotamine (DHE) that reactivate NR4A expression and regulate NR4A-dependent gene signatures. We show that DHE induces NR4A expression via recruitment of the super elongation complex to enable elongation of NR4A promoter paused RNA polymerase II. Finally, DHE exhibits AML selective NR4A-dependent anti-leukemic activity in cytogenetically distinct human AML cells in vitro and delays AML progression in mice revealing its potential as a novel therapeutic agent in AML.


Asunto(s)
Dihidroergotamina/farmacología , Sistemas de Liberación de Medicamentos/métodos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Leucemia Mieloide Aguda/tratamiento farmacológico , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Vasoconstrictores/farmacología , Animales , Apoptosis , Proliferación Celular , Epigénesis Genética , Femenino , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Transcriptoma , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
3.
PLoS One ; 13(3): e0194913, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29590203

RESUMEN

The tumor suppressor gene p53 is frequently mutated in human breast cancer and is a marker for poor prognosis and resistance to chemotherapy. Transplantation of p53 null mouse mammary epithelium into syngeneic wild-type mice leads to normal mammary gland development followed by spontaneous mammary tumors that recapitulate many of the phenotypic, molecular and genetic features of human breast cancer. Transient exposure of p53 null mice to the anti-estrogen, tamoxifen leads to sustained and robust protection against tumor development. However the mechanism underlying this anti-tumor activity remains poorly understood. Here we demonstrate that transient exposure to tamoxifen leads to a reduction in mammary ductal side-branching and epithelial cell proliferation after tamoxifen withdrawal. Global gene expression analysis showed that transient tamoxifen exposure leads to persistent changes in the expression of a subset of estrogen regulated gene signatures in mammary epithelial cells (MECs). Among these was the protein tyrosine phosphatase, non-receptor type 5 (Ptpn5). We show that Ptpn5 is a novel tamoxifen regulated target gene which is upregulated in MECs after transient tamoxifen exposure and displays tumor suppressor activity in human breast cancer cells. Further, PTPN5 expression is strongly associated with good clinical outcome in tamoxifen treated human breast cancer patients suggesting that PTPN5 may represent a novel biomarker of tamoxifen response in human breast cancer.


Asunto(s)
Neoplasias de la Mama/prevención & control , Transformación Celular Neoplásica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Mamarias Experimentales/prevención & control , Tamoxifeno/farmacología , Transcriptoma/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo , Animales , Antineoplásicos Hormonales/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Células Cultivadas , Femenino , Genes Supresores de Tumor , Humanos , Glándulas Mamarias Animales/efectos de los fármacos , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/genética , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Receptores de Estrógenos/metabolismo , Proteína p53 Supresora de Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Blood ; 131(10): 1081-1093, 2018 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-29343483

RESUMEN

Members of the NR4A subfamily of nuclear receptors have complex, overlapping roles during hematopoietic cell development and also function as tumor suppressors of hematologic malignancies. We previously identified NR4A1 and NR4A3 (NR4A1/3) as functionally redundant suppressors of acute myeloid leukemia (AML) development. However, their role in hematopoietic stem cell (HSC) homeostasis remains to be disclosed. Using a conditional Nr4a1/Nr4a3 knockout mouse (CDKO), we show that codepletion of NR4A1/3 promotes acute changes in HSC homeostasis including loss of HSC quiescence, accumulation of oxidative stress, and DNA damage while maintaining stem cell regenerative and differentiation capacity. Molecular profiling of CDKO HSCs revealed widespread upregulation of genetic programs governing cell cycle and inflammation and an aberrant activation of the interferon and NF-κB signaling pathways in the absence of stimuli. Mechanistically, we demonstrate that NR4A1/3 restrict HSC proliferation in part through activation of a C/EBPα-driven antiproliferative network by directly binding to a hematopoietic-specific Cebpa enhancer and activating Cebpa transcription. In addition, NR4A1/3 occupy the regulatory regions of NF-κB-regulated inflammatory cytokines, antagonizing the activation of NF-κB signaling. Taken together, our results reveal a novel coordinate control of HSC quiescence by NR4A1/3 through direct activation of C/EBPα and suppression of activation of NF-κB-driven proliferative inflammatory responses.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/inmunología , Proliferación Celular , Proteínas de Unión al ADN/inmunología , Células Madre Hematopoyéticas/inmunología , Proteínas del Tejido Nervioso/inmunología , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/inmunología , Receptores de Esteroides/inmunología , Receptores de Hormona Tiroidea/inmunología , Transducción de Señal/inmunología , Animales , Proteínas Potenciadoras de Unión a CCAAT/genética , Daño del ADN , Proteínas de Unión al ADN/genética , Elementos de Facilitación Genéticos/inmunología , Células Madre Hematopoyéticas/patología , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Estrés Oxidativo/genética , Estrés Oxidativo/inmunología , Receptores de Esteroides/genética , Receptores de Hormona Tiroidea/genética , Transducción de Señal/genética , Transcripción Genética/inmunología
5.
Oncotarget ; 8(33): 54364-54377, 2017 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-28903348

RESUMEN

The nuclear receptor (NR) superfamily contains hormone-inducible transcription factors that regulate many physiological and pathological processes through regulating gene expression. NR4A1 is an NR family member that still does not have an identified endogenous ligand, and its role in cancer is also currently unclear and controversial. In this study, we aimed to define the expression profiles and specific role of NR4A1 in the highly malignant triple-negative breast cancer (TNBC), which still lacks available targeted therapies. Bioinformatic analysis revealed a decrease of NR4A1 mRNA expression in human TNBC samples. Semi-quantitative analysis of NR4A1 protein expression by immunohistochemistry also identified a progressive NR4A1 reduction during the development of mouse basal-like mammary tumors and a significant NR4A1 downregulation in human TNBC samples. Furthermore, the expression levels of NR4A1 in human TNBC were negatively associated with tumor stage, lymph node metastasis and disease recurrence. Moreover, ectopic expression of NR4A1 in MDA-MB-231, a TNBC cell line with little endogenous NR4A1, inhibited the proliferation, viability, migration and invasion of these cells, and these inhibitions were associated with an attenuated JNK1-AP-1-cyclin D1 pathway. NR4A1 expression also largely suppressed the growth and metastasis of these cell-derived tumors in mice. These results demonstrate that NR4A1 is downregulated in TNBC and restoration of NR4A1 expression inhibits TNBC growth and metastasis, suggesting that NR4A1 is a tumor suppressor in TNBC.

6.
PLoS One ; 11(3): e0150450, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26938745

RESUMEN

Members of the NR4A subfamily of orphan nuclear receptors regulate cell fate decisions via both genomic and non-genomic mechanisms in a cell and tissue selective manner. NR4As play a key role in maintenance of hematopoietic stem cell homeostasis and are critical tumor suppressors of acute myeloid leukemia (AML). Expression of NR4As is broadly silenced in leukemia initiating cell enriched populations from human patients relative to normal hematopoietic stem/progenitor cells. Rescue of NR4A expression in human AML cells inhibits proliferation and reprograms AML gene signatures via transcriptional mechanisms that remain to be elucidated. By intersecting an acutely regulated NR4A1 dependent transcriptional profile with genome wide NR4A binding distribution, we now identify an NR4A targetome of 685 genes that are directly regulated by NR4A1. We show that NR4As regulate gene transcription primarily through interaction with distal enhancers that are co-enriched for NR4A1 and ETS transcription factor motifs. Using a subset of NR4A activated genes, we demonstrate that the ETS factors ERG and FLI-1 are required for activation of NR4A bound enhancers and NR4A target gene induction. NR4A1 dependent recruitment of ERG and FLI-1 promotes binding of p300 histone acetyltransferase to epigenetically activate NR4A bound enhancers via acetylation at histone H3K27. These findings disclose novel epigenetic mechanisms by which NR4As and ETS factors cooperate to drive NR4A dependent gene transcription in human AML cells.


Asunto(s)
Epigénesis Genética , Leucemia Mieloide Aguda/genética , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Proteína Proto-Oncogénica c-ets-1/genética , Secuencias de Aminoácidos , Linaje de la Célula , Proliferación Celular , Inmunoprecipitación de Cromatina , Mapeo Cromosómico , Biología Computacional , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/citología , Histonas/química , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Plásmidos/metabolismo , Unión Proteica , Proteína Proto-Oncogénica c-fli-1/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Transcripción Genética , Factores de Transcripción p300-CBP/metabolismo
7.
eNeuro ; 2(4)2015.
Artículo en Inglés | MEDLINE | ID: mdl-26465008

RESUMEN

Estrogens act in brain to profoundly influence neurogenesis, sexual differentiation, neuroprotection, cognition, energy homeostasis, and female reproductive behavior and physiology through a variety of mechanisms, including the induction of progestin receptors (PRs). PRs are expressed as two isoforms, PR-A and PR-B, that have distinct functions in physiology and behavior. Because these PR isoforms cannot be distinguished using cellular resolution techniques, the present study used isoform-specific null mutant mice that lack PR-A or PR-B for the first time to investigate whether 17ß-estradiol benzoate (EB) regulates the differential expression of the PR isoforms in the ventromedial nucleus of the hypothalamus (VMN), arcuate nucleus, and medial preoptic area, brain regions that are rich in EB-induced PRs. Interestingly, EB induced more PR-A than PR-B in all three brain regions, suggesting that PR-A is the predominant isoform in these regions. Given that steroid receptor coactivator (SRC)-1 and SRC-2 are important in estrogen receptor (ER)-dependent transcription in brain, including PR induction, we tested whether the expression of these coactivators was correlated with PR isoform expression. The majority of EB-induced PR cells expressed both SRC-1 and SRC-2 in the three brain regions of all genotypes. Interestingly, the intensity of PR-A immunoreactivity correlated with SRC-2 expression in the VMN, providing a potential mechanism for selective ER-mediated transactivation of PR-A over PR-B in a brain region-specific manner. In summary, these novel findings indicate that estrogens differentially regulate PR-A and PR-B expression in the female hypothalamus, and provide a mechanism by which steroid action in brain can selectively modulate behavior and physiology.

8.
Mol Cell Biol ; 35(7): 1125-38, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25605333

RESUMEN

We previously showed that the orphan nuclear receptor Nur77 (Nr4a1) plays an important role in the regulation of glucose homeostasis and oxidative metabolism in skeletal muscle. Here, we show using both gain- and loss-of-function models that Nur77 is also a regulator of muscle growth in mice. Transgenic expression of Nur77 in skeletal muscle in mice led to increases in myofiber size. Conversely, mice with global or muscle-specific deficiency in Nur77 exhibited reduced muscle mass and myofiber size. In contrast to Nur77 deficiency, deletion of the highly related nuclear receptor NOR1 (Nr4a3) had minimal effect on muscle mass and myofiber size. We further show that Nur77 mediates its effects on muscle size by orchestrating transcriptional programs that favor muscle growth, including the induction of insulin-like growth factor 1 (IGF1), as well as concomitant downregulation of growth-inhibitory genes, including myostatin, Fbxo32 (MAFbx), and Trim63 (MuRF1). Nur77-mediated increase in IGF1 led to activation of the Akt-mTOR-S6K cascade and the inhibition of FoxO3a activity. The dependence of Nur77 on IGF1 was recapitulated in primary myoblasts, establishing this as a cell-autonomous effect. Collectively, our findings identify Nur77 as a novel regulator of myofiber size and a potential transcriptional link between cellular metabolism and muscle growth.


Asunto(s)
Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/crecimiento & desarrollo , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Animales , Células Cultivadas , Femenino , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fibras Musculares Esqueléticas/ultraestructura , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Mutación , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Proteínas Ligasas SKP Cullina F-box/genética , Proteínas Ligasas SKP Cullina F-box/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
9.
PLoS One ; 9(2): e89311, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24586680

RESUMEN

Nuclear receptor 4a3 (Nr4a3) is a transcription factor implicated in various settings such as vascular biology and inflammation. We have recently shown that mast cells dramatically upregulate Nuclear receptor 4a3 upon activation, and here we investigated the functional impact of Nuclear receptor 4a3 on mast cell responses. We show that Nuclear receptor 4a3 is involved in the regulation of cytokine/chemokine secretion in mast cells following activation via the high affinity IgE receptor. Moreover, Nuclear receptor 4a3 negatively affects the transcript and protein levels of mast cell tryptase as well as the mast cell's responsiveness to allergen. Together, these findings identify Nuclear receptor 4a3 as a novel regulator of mast cell function.


Asunto(s)
Gránulos Citoplasmáticos/metabolismo , Proteínas de Unión al ADN/fisiología , Mastocitos/metabolismo , Proteínas del Tejido Nervioso/fisiología , Receptores de Esteroides/fisiología , Receptores de Hormona Tiroidea/fisiología , Animales , Apoptosis , Western Blotting , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Proliferación Celular , Células Cultivadas , Quimiocinas/genética , Quimiocinas/metabolismo , Citocinas/genética , Citocinas/metabolismo , Gránulos Citoplasmáticos/efectos de los fármacos , Gránulos Citoplasmáticos/inmunología , Inmunoglobulina E/farmacología , Mastocitos/citología , Mastocitos/efectos de los fármacos , Mastocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/genética , FN-kappa B/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de IgE/genética , Receptores de IgE/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Triptasas/genética , Triptasas/metabolismo
10.
Mol Endocrinol ; 27(10): 1743-61, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23979845

RESUMEN

Progesterone (P4)-activated progesterone receptors (PRs) play an essential role in driving pregnancy-associated mammary ductal side-branching morphogenesis and alveologenesis. However, the global cistromic and transcriptome responses that are required to elicit P4-dependent branching morphogenesis have not been elucidated. By combining chromatin immunoprecipitation followed by deep sequencing to identify genome-wide PR-binding sites in PR-positive luminal epithelial cells with global gene expression signatures acutely regulated by PRs in the mammary gland, we have identified a mammary epithelial PR targetome associated with active P4-dependent branching morphogenesis in vivo. We demonstrate that P4-induced side-branching is initiated by epithelial cell rearrangement into a multilayered epithelium that sprouts laterally from quiescent ducts via a mechanism requiring P4-dependent activation of Rac-GTPase signaling. We identify effectors of Rac-GTPases as direct transcriptional targets of PRs, and we demonstrate that disruption of the P4-activated Rac-GTPase signaling axis is sufficient to eliminate P4-dependent side-branching. Our data reveal that the molecular mediators of P4-dependent ductal side-branching overlap with those implicated in breast cancer.


Asunto(s)
Glándulas Mamarias Animales/metabolismo , Morfogénesis , Progesterona/fisiología , Receptores de Progesterona/genética , Animales , Epitelio/metabolismo , Estradiol/fisiología , Femenino , Regulación de la Expresión Génica , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/crecimiento & desarrollo , Ratones de la Cepa 129 , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Progesterona/metabolismo , Transducción de Señal , Transcriptoma , Proteínas de Unión al GTP rac/genética , Proteínas de Unión al GTP rac/metabolismo
11.
J Lipid Res ; 54(3): 806-815, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23288947

RESUMEN

The formation of the atherosclerotic lesion is a complex process influenced by an array of inflammatory and lipid metabolism pathways. We previously demonstrated that NR4A nuclear receptors are highly induced in macrophages in response to inflammatory stimuli and modulate the expression of genes linked to inflammation in vitro. Here we used mouse genetic models to assess the impact of NR4A expression on atherosclerosis development and macrophage polarization. Transplantation of wild-type, Nur77⁻/⁻, or Nor1⁻/⁻ null hematopoetic precursors into LDL receptor (LDLR)⁻/⁻ recipient mice led to comparable development of atherosclerotic lesions after high-cholesterol diet. We also observed comparable induction of genes linked to M1 and M2 responses in wild-type and Nur77-null macrophages in response to lipopolysaccharides and interleukin (IL)-4, respectively. In contrast, activation of the nuclear receptor liver X receptor (LXR) strongly suppressed M1 responses, and ablation of signal transductor and activator of transcription 6 (STAT6) strongly suppressed M2 responses. Recent studies have suggested that alterations in levels of Ly6C(lo) monocytes may be a contributor to inflammation and atherosclerosis. In our study, loss of Nur77, but not Nor1, was associated with decreased abundance of Ly6C(lo) monocytes, but this change was not correlated with atherosclerotic lesion development. Collectively, our results suggest that alterations in the Ly6C(lo) monocyte population and bone marrow NR4A expression do not play dominant roles in macrophage polarization or the development of atherosclerosis in mice.


Asunto(s)
Aterosclerosis/metabolismo , Macrófagos/metabolismo , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Animales , Aterosclerosis/genética , Citometría de Flujo , Masculino , Ratones , Ratones Noqueados , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
12.
Blood ; 117(9): 2681-90, 2011 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-21205929

RESUMEN

The NR4A subfamily of nuclear receptors (NR4A1, NR4A2, and NR4A3) function as transcription factors that transduce diverse extracellular signals into altered gene transcription to coordinate apoptosis, proliferation, cell cycle arrest, and DNA repair. We previously discovered that 2 of these receptors, NR4A1 and NR4A3, are potent tumor suppressors of acute myeloid leukemia (AML); they are silenced in human AML, and abrogation of both genes in mice leads to rapid postnatal development of AML. Reduced expression of NR4As is also a common feature of myelodysplastic syndromes (MDSs). Here we show that reduced gene dosage of NR4A1 and NR4A3 in hypoallelic (NR4A1(+/-)NR4A3(-/-) or NR4A1(-/-)NR4A3(+/-)) mice below a critical threshold leads to a chronic myeloid malignancy that closely recapitulates the pathologic features of mixed myelodysplastic/myeloproliferative neoplasms (MDS/MPNs) with progression to AML in rare cases. Enhanced proliferation and excessive apoptosis of hematopoietic stem cells and myeloid progenitors, together with elevated DNA damage, contribute to MDS/MPN disease. We identify the myeloid tumor suppressor genes Egr1 and JunB and the DNA damage checkpoint kinase, polo-like kinase 2 (Plk2) as deregulated genes whose disrupted signaling probably contributes to MDS/MPN. These mice provide a novel model to elucidate the molecular pathogenesis of MDS/MPN and for therapeutic evaluation.


Asunto(s)
Proteínas de Unión al ADN/genética , Dosificación de Gen/genética , Síndromes Mielodisplásicos/genética , Trastornos Mieloproliferativos/genética , Proteínas del Tejido Nervioso/genética , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Receptores de Esteroides/genética , Receptores de Hormona Tiroidea/genética , Alelos , Animales , Apoptosis , Compartimento Celular , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Daño del ADN , Progresión de la Enfermedad , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Humanos , Leucemia Mieloide Aguda/patología , Ratones , Ratones Endogámicos C57BL , Síndromes Mielodisplásicos/patología , Células Progenitoras Mieloides/patología , Trastornos Mieloproliferativos/patología , Fenotipo , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas
13.
Nat Cell Biol ; 12(12): 1213-9, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21076412

RESUMEN

Successful haematopoiesis requires long-term retention of haematopoietic stem cells (HSCs) in a quiescent state. The transcriptional regulation of stem cell quiescence, especially by factors with specific functions in HSCs, is only beginning to be understood. Here, we demonstrate that Nurr1, a nuclear receptor transcription factor, has such a regulatory role. Overexpression of Nurr1 drives early haematopoietic progenitors into quiescence. When stem cells overexpressing Nurr1 are transplanted into lethally irradiated mice, they localize to the bone marrow, but do not contribute to regeneration of the blood system. Furthermore, the loss of only one allele of Nurr1 is sufficient to induce HSCs to enter the cell cycle and proliferate. Molecular analysis revealed an association between Nurr1 overexpression and upregulation of the cell-cycle inhibitor p18 (also known as INK4C), suggesting a mechanism by which Nurr1 could regulate HSC quiescence. Our findings provide critical insight into the transcriptional control mechanisms that determine whether HSCs remain dormant or enter the cell cycle and begin to proliferate.


Asunto(s)
Proliferación Celular , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Animales , Médula Ósea , Ciclo Celular , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/metabolismo , Regulación de la Expresión Génica , Células Madre Hematopoyéticas , Ratones , Ratones Endogámicos C57BL , Transcripción Genética
14.
Handb Exp Pharmacol ; (198): 37-44, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20839085

RESUMEN

The steroid hormone, progesterone, plays a critical role in the regulation of female ovulation. The physiological effects of progesterone are mediated by two nuclear receptor transcription factors, PR-A and PR-B, which are produced from a single gene and upon binding progesterone regulate the expression of specific gene networks in reproductive tissues. Both null mutation of the PR gene to delete both receptor proteins and selective disruption of the PR-A isoform lead to a failure of ovulation due to disabled follicular rupture in response to gonadotrophin stimulation. Recent studies have revealed that the LH stimulus that triggers ovulation is transduced by PRs residing in mural granulosa cells that induce expression of paracrine signals that interact with cumulus cells to control cumulus matrix function and expansion to facilitate follicular rupture.


Asunto(s)
Ovulación/efectos de los fármacos , Receptores de Progesterona/efectos de los fármacos , Receptores de Progesterona/fisiología , Animales , Células del Cúmulo/efectos de los fármacos , Femenino , Humanos , Hormona Luteinizante/farmacología , Folículo Ovárico/efectos de los fármacos , Comunicación Paracrina/fisiología , Elementos Reguladores de la Transcripción/fisiología , Transducción de Señal/efectos de los fármacos
15.
Circ Res ; 107(4): 501-11, 2010 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-20558821

RESUMEN

RATIONALE: The orphan nuclear receptor NOR1 is a member of the evolutionary highly conserved and ligand-independent NR4A subfamily of the nuclear hormone receptor superfamily. Members of this subfamily have been characterized as early response genes regulating essential biological processes including inflammation and proliferation; however, the role of NOR1 in atherosclerosis remains unknown. OBJECTIVE: The goal of the present study was to determine the causal contribution of NOR1 to atherosclerosis development and to identify the mechanism by which this nuclear receptor participates in the disease process. METHODS AND RESULTS: In the present study, we demonstrate expression of NOR1 in endothelial cells of human atherosclerotic lesions. In response to inflammatory stimuli, NOR1 expression is rapidly induced in endothelial cells through a nuclear factor kappaB-dependent transactivation of the NOR1 promoter. Overexpression of NOR1 in human endothelial cells increased the expression of vascular cell adhesion molecule (VCAM)-1 and intercellular adhesion molecule-1, whereas NOR1 deficiency altered adhesion molecule expression in response to inflammatory stimuli. Transient transfection experiments and chromatin immunoprecipitation assays revealed that NOR1 induces VCAM-1 promoter activity by binding to a canonical response element for NR4A receptors in the VCAM-1 promoter. Further functional studies confirmed that NOR1 mediates monocyte adhesion by inducing VCAM-1 and intercellular adhesion molecule-1 expression in endothelial cells. Finally, we demonstrate that NOR1 deficiency reduces hypercholesterolemia-induced atherosclerosis formation in apoE(-/-) mice by decreasing the macrophage content of the lesion. CONCLUSIONS: In concert, these studies identify a novel pathway underlying monocyte adhesion and establish that NOR1 serves a previously unrecognized atherogenic role in mice by positively regulating monocyte recruitment to the vascular wall.


Asunto(s)
Aterosclerosis/metabolismo , Proteínas de Unión al ADN/deficiencia , Monocitos/fisiología , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/deficiencia , Receptores de Esteroides/deficiencia , Receptores de Hormona Tiroidea/deficiencia , Animales , Aterosclerosis/patología , Aterosclerosis/prevención & control , Adhesión Celular/fisiología , Células Cultivadas , Vasos Coronarios/metabolismo , Vasos Coronarios/patología , Proteínas de Unión al ADN/metabolismo , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Receptores de Esteroides/metabolismo , Receptores de Hormona Tiroidea/metabolismo , Elementos de Respuesta/genética , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo
16.
Circulation ; 119(4): 577-86, 2009 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-19153266

RESUMEN

BACKGROUND: The neuron-derived orphan receptor-1 (NOR1) belongs to the evolutionary highly conserved and most ancient NR4A subfamily of the nuclear hormone receptor superfamily. Members of this subfamily function as early-response genes regulating key cellular processes, including proliferation, differentiation, and survival. Although NOR1 has previously been demonstrated to be required for smooth muscle cell proliferation in vitro, the role of this nuclear receptor for the proliferative response underlying neointima formation and target genes trans-activated by NOR1 remain to be defined. METHODS AND RESULTS: Using a model of guidewire-induced arterial injury, we demonstrate decreased neointima formation in NOR1(-/-) mice compared with wild-type mice. In vitro, NOR1-deficient smooth muscle cells exhibit decreased proliferation as a result of a G(1)-->S phase arrest of the cell cycle and increased apoptosis in response to serum deprivation. NOR1 deficiency alters phosphorylation of the retinoblastoma protein by preventing mitogen-induced cyclin D1 and D2 expression. Conversely, overexpression of NOR1 induces cyclin D1 expression and the transcriptional activity of the cyclin D1 promoter in transient reporter assays. Gel shift and chromatin immunoprecipitation assays identified a putative response element for NR4A receptors in the cyclin D1 promoter, to which NOR1 is recruited in response to mitogenic stimulation. Finally, we provide evidence that these observations are applicable in vivo by demonstrating decreased cyclin D1 expression during neointima formation in NOR1-deficient mice. CONCLUSIONS: These experiments characterize cyclin D1 as an NOR1-regulated target gene in smooth muscle cells and demonstrate that NOR1 deficiency decreases neointima formation in response to vascular injury.


Asunto(s)
Proteínas de Unión al ADN/genética , Músculo Liso Vascular/lesiones , Músculo Liso Vascular/fisiología , Receptores de Esteroides/genética , Cicatrización de Heridas/fisiología , Animales , Aorta/citología , Apoptosis/fisiología , División Celular/fisiología , Supervivencia Celular/fisiología , Células Cultivadas , Vasos Coronarios/citología , Ciclina D1/genética , Ciclina D2 , Ciclinas/genética , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción E2F/metabolismo , Expresión Génica/fisiología , Humanos , Ratones , Ratones Mutantes , Músculo Liso Vascular/citología , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares , Fosforilación/fisiología , Regiones Promotoras Genéticas/fisiología , Ratas , Receptores de Esteroides/metabolismo , Proteína de Retinoblastoma/metabolismo , Túnica Íntima/citología , Túnica Íntima/lesiones , Túnica Íntima/fisiología
17.
Am J Pathol ; 172(4): 1019-29, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18321995

RESUMEN

Lactoferrin (LF) is an iron-binding protein found in milk, mucosal secretions, and the secondary granules of neutrophils in which it is considered to be an important factor in the innate immune response against microbial infections. Moreover, LF deficiency in the secondary granules of neutrophils has long been speculated to contribute directly to the hypersusceptibility of specific granule deficiency (SGD) patients to severe, life-threatening bacterial infections. However, the exact physiological significance of LF in neutrophil-mediated host defense mechanisms remains controversial and has not yet been clearly established in vivo using relevant animal models. In this study, we used lactoferrin knockout (LFKO) mice to directly address the selective role of LF in the host defense response of neutrophils and to determine its contribution, if any, to the phenotype of SGD. Neutrophil maturation, migration, phagocytosis, granule release, and antimicrobial response to bacterial challenge were unaffected in LFKO mice. Interestingly, a stimulus-dependent defect in the oxidative burst response of LFKO neutrophils was observed in that normal activation was seen in response to opsonized bacteria whereas an impaired response was evident after phorbol myristate-13-acetate stimulation. Taken together, these results indicate that although LF deficiency alone is not a primary cause of the defects associated with SGD, this protein does play an immunomodulatory role in the oxidative burst response of neutrophils.


Asunto(s)
Lactoferrina/deficiencia , Neutrófilos/inmunología , Neutrófilos/metabolismo , Estallido Respiratorio/inmunología , Animales , Western Blotting , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Susceptibilidad a Enfermedades/inmunología , Susceptibilidad a Enfermedades/microbiología , Relación Dosis-Respuesta Inmunológica , Leucocitos/efectos de los fármacos , Leucocitos/microbiología , Pulmón/efectos de los fármacos , Pulmón/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , NADPH Oxidasas/metabolismo , Neutrófilos/enzimología , Neutrófilos/microbiología , Peroxidasa/metabolismo , Fagocitosis/efectos de los fármacos , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/fisiología , Estallido Respiratorio/efectos de los fármacos , Vesículas Secretoras/metabolismo , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/fisiología , Acetato de Tetradecanoilforbol/farmacología
18.
Cancer Res ; 67(24): 12026-33, 2007 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-18089834

RESUMEN

The use of agents to prevent the onset of and/or the progression to breast cancer has the potential to lower breast cancer risk. We have previously shown that the tumor-suppressor gene p53 is a potential mediator of hormone (estrogen/progesterone)-induced protection against chemical carcinogen-induced mammary carcinogenesis in animal models. Here, we show for the first time a breast cancer-protective effect of chloroquine in an animal model. Chloroquine significantly reduced the incidence of N-methyl-N-nitrosourea-induced mammary tumors in our animal model similar to estrogen/progesterone treatment. No protection was seen in our BALB/c p53-null mammary epithelium model, indicating a p53 dependency for the chloroquine effect. Using a human nontumorigenic mammary gland epithelial cell line, MCF10A, we confirm that in the absence of detectable DNA damage, chloroquine activates the tumor-suppressor p53 and the p53 downstream target gene p21, resulting in G(1) cell cycle arrest. p53 activation occurs at a posttranslational level via chloroquine-dependent phosphorylation of the checkpoint protein kinase, ataxia telangiectasia-mutated (ATM), leading to ATM-dependent phosphorylation of p53. In primary mammary gland epithelial cells isolated from p53-null mice, chloroquine does not induce G(1) cell cycle arrest compared with cells isolated from wild-type mice, also indicating a p53 dependency. Our results indicate that a short prior exposure to chloroquine may have a preventative application for mammary carcinogenesis.


Asunto(s)
Proteínas de Ciclo Celular/genética , Cloroquina/farmacología , Proteínas de Unión al ADN/genética , Genes p53 , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/prevención & control , Proteínas Serina-Treonina Quinasas/genética , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética , Proteínas Supresoras de Tumor/genética , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Mama/citología , Mama/fisiología , Proteínas de Ciclo Celular/fisiología , Proteínas de Unión al ADN/fisiología , Resistencia a Antineoplásicos , Células Epiteliales/citología , Células Epiteliales/fisiología , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Supresoras de Tumor/fisiología
19.
Nat Med ; 13(6): 730-5, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17515897

RESUMEN

Nur77 (NR4A1) and Nor-1 (NR4A3) are highly homologous orphan nuclear receptors that regulate the transcription of overlapping target genes. The transcriptional activity of both proteins is regulated in a ligand-independent manner by cell- and stimulus-specific gene induction and protein phosphorylation. Nor-1 and Nur77 have been implicated in a variety of cellular processes, including the transduction of hormonal, inflammatory, mitogenic, apoptotic and differentiative signals. Cellular responses to these proteins suggest that they may function as homeostatic regulators of proliferation, apoptosis and differentiation, and thus may regulate cellular susceptibility to tumorigenesis. Their physiological functions, however, remain poorly understood. Here we describe a previously unsuspected function of Nor-1 and Nur77-as critical tumor suppressors of myeloid leukemogenesis. The abrogation of these proteins in mice led to rapidly lethal acute myeloid leukemia (AML), involving abnormal expansion of hematopoietic stem cells (HSCs) and myeloid progenitors, decreased expression of the AP-1 transcription factors JunB and c-Jun and defective extrinsic apoptotic (Fas-L and TRAIL) signaling. We found that downregulation of NR4A3 ( NOR-1 ) and NR4A1 ( NUR77 ) was a common feature in leukemic blasts from human AML patients, irrespective of karyotype. Thus Nor-1 and Nur77 may provide potential targets for therapeutic intervention in AML.


Asunto(s)
Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/fisiología , Leucemia Mieloide/genética , Leucemia Mieloide/metabolismo , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/fisiología , Receptores Citoplasmáticos y Nucleares/deficiencia , Receptores Citoplasmáticos y Nucleares/fisiología , Receptores de Esteroides/deficiencia , Receptores de Esteroides/fisiología , Receptores de Hormona Tiroidea/deficiencia , Receptores de Hormona Tiroidea/fisiología , Factores de Transcripción/deficiencia , Factores de Transcripción/fisiología , Enfermedad Aguda , Animales , Crisis Blástica/genética , Crisis Blástica/patología , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Regulación hacia Abajo/genética , Humanos , Leucemia Mieloide/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Receptores Citoplasmáticos y Nucleares/biosíntesis , Receptores Citoplasmáticos y Nucleares/genética , Receptores de Esteroides/antagonistas & inhibidores , Receptores de Esteroides/biosíntesis , Receptores de Esteroides/genética , Receptores de Hormona Tiroidea/antagonistas & inhibidores , Receptores de Hormona Tiroidea/biosíntesis , Receptores de Hormona Tiroidea/genética , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...