Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros












Intervalo de año de publicación
1.
Hepatol Commun ; 8(8)2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-39023343

RESUMEN

BACKGROUND: When massive necrosis occurs in acute liver failure (ALF), rapid expansion of HSCs called liver progenitor cells (LPCs) in a process called ductular reaction is required for survival. The underlying mechanisms governing this process are not entirely known to date. In ALF, high levels of retinoic acid (RA), a molecule known for its pleiotropic roles in embryonic development, are secreted by activated HSCs. We hypothesized that RA plays a key role in ductular reaction during ALF. METHODS: RNAseq was performed to identify molecular signaling pathways affected by all-trans retinoid acid (atRA) treatment in HepaRG LPCs. Functional assays were performed in HepaRG cells treated with atRA or cocultured with LX-2 cells and in the liver tissue of patients suffering from ALF. RESULTS: Under ALF conditions, activated HSCs secreted RA, inducing RARα nuclear translocation in LPCs. RNAseq data and investigations in HepaRG cells revealed that atRA treatment activated the WNT-ß-Catenin pathway, enhanced stemness genes (SOX9, AFP, and others), increased energy storage, and elevated the expression of ATP-binding cassette transporters in a RARα nuclear translocation-dependent manner. Further, atRA treatment-induced pathways were confirmed in a coculture system of HepaRG with LX-2 cells. Patients suffering from ALF who displayed RARα nuclear translocation in the LPCs had significantly better MELD scores than those without. CONCLUSIONS: During ALF, RA secreted by activated HSCs promotes LPC activation, a prerequisite for subsequent LPC-mediated liver regeneration.


Asunto(s)
Fallo Hepático Agudo , Células Madre , Tretinoina , Humanos , Tretinoina/farmacología , Células Madre/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos , Hígado/efectos de los fármacos , Receptor alfa de Ácido Retinoico/genética , Receptor alfa de Ácido Retinoico/metabolismo , Técnicas de Cocultivo , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/metabolismo
2.
Sci Total Environ ; 947: 174717, 2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-38997027

RESUMEN

Retinoic acid receptors (RARs) are known as crucial endocrine receptors that could mediate a broad diversity of biological processes. However, the data on endocrine disrupting effects of emerging chemicals by targeting RAR (ant)agonism are far from sufficient. Herein, we investigated the RARα agonistic or antagonistic activities for 75 emerging chemicals of concern, and explored their interactions with this receptor. A recombinant two-hybrid yeast assay was used to examine the RARα activities of the test chemicals, wherein 7 showed effects of RARα agonism and 54 exerted potentials of RARα antagonism. The representative chemicals with RARα agonistic activities, i.e. 4-hydroxylphenol (4-HP) and bisphenol AF (BPAF), significantly increased the mRNA levels of CRABP2 and CYP26A1, while 4 select chemicals with RARα antagonistic potentials, including bisphenol A (BPA), tetrabromobisphenol A (TBBPA), 4-tert-octylphenol (4-t-OP), and 4-n-nonylphenol (4-n-NP), conversely decreased the transcriptional levels of the test genes. The in silico molecular docking analysis using 3 different approaches further confirmed the substantial binding between the chemicals with RARα activities and this nuclear receptor protein. This work highlights the promising strategy for screening endocrine-disrupting effects of emerging chemicals of concern by targeting RARα (ant)agonism.


Asunto(s)
Disruptores Endocrinos , Receptor alfa de Ácido Retinoico , Xenobióticos , Receptor alfa de Ácido Retinoico/metabolismo , Receptor alfa de Ácido Retinoico/genética , Humanos , Simulación del Acoplamiento Molecular , Simulación por Computador , Receptores de Ácido Retinoico/metabolismo
3.
Mod Pathol ; 37(7): 100509, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38704030

RESUMEN

Acute promyelocytic leukemia (APL) with variant RARA translocation is linked to over 15 partner genes. Recent publications encompassing 6 cases have expanded the spectrum of RARA partners to torque teno mini virus (TTMV). This entity is likely underrecognized due to the lack of clinician and pathologist familiarity, inability to detect the fusion using routine testing modalities, and informatic challenges in its recognition within next-generation sequencing (NGS) data. We describe a clinicopathologic approach and provide the necessary tools to screen and diagnose APL with TTMV::RARA using existing clinical DNA- or RNA-based NGS assays, which led to the identification of 4 cases, all without other known cytogenetic/molecular drivers. One was identified prospectively and 3 retrospectively, including 2 from custom automated screening of multiple data sets (50,257 cases of hematopoietic malignancy, including 4809 acute myeloid leukemia/myeloid sarcoma/APL cases). Two cases presented as myeloid sarcoma, including 1 with multiple relapses after acute myeloid leukemia-type chemotherapy and hematopoietic stem cell transplant. Two cases presented as leukemia, had a poor response to induction chemotherapy, but achieved remission upon reinduction (including all-trans retinoic acid in 1 case) and subsequent hematopoietic stem cell transplant. Neoplastic cells demonstrated features of APL including frequent azurophilic granules and dim/absent CD34 and HLA-DR expression. RARA rearrangement was not detected by karyotype or fluorescent in situ hybridization. Custom analysis of NGS fusion panel data identified TTMV::RARA rearrangements and, in the prospectively identified case, facilitated monitoring in sequential bone marrow samples. APL with TTMV::RARA is a rare leukemia with a high rate of treatment failure in described cases. The diagnosis should be considered in leukemias with features of APL that lack detectable RARA fusions and other drivers, and may be confirmed by appropriate NGS tests with custom informatics. Incorporation of all-trans retinoic acid may have a role in treatment but requires accurate recognition of the fusion for appropriate classification as APL.


Asunto(s)
Leucemia Promielocítica Aguda , Proteínas de Fusión Oncogénica , Receptor alfa de Ácido Retinoico , Torque teno virus , Humanos , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/diagnóstico , Receptor alfa de Ácido Retinoico/genética , Masculino , Torque teno virus/genética , Proteínas de Fusión Oncogénica/genética , Femenino , Adulto , Persona de Mediana Edad , Secuenciación de Nucleótidos de Alto Rendimiento
4.
FEBS J ; 291(15): 3521-3538, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38708519

RESUMEN

HER2-enriched (HER2+) breast cancers express high levels of the growth-promoting HER2 protein. Although these cancers are treated with the HER2-targeted drug, trastuzumab, resistance to treatment is common. Retinoic acid (RA) is an anti-cancer agent that has been successfully used for the treatment of leukemia and holds promise for the treatment of solid cancers, including breast cancer. The HER2 gene is frequently co-amplified with RARA, a key determinant of RA sensitivity in breast cancers. It seems surprising, therefore, that HER2+ breast cancers are refractory to RA treatment. Here, we show that MYC mediates RA resistance by suppressing the expression of cellular retinoic acid binding protein 2 (CRABP2), resulting in RARα inactivation. CRABP2 is an intracellular RA transporter that delivers RA to the nuclear receptor RARα for its activation. Our results indicate that response to RA is enhanced by MYC depletion in HER2+ breast cancer cells and that RA treatment enhances trastuzumab responsiveness. Our findings support the use of RA and trastuzumab for the treatment of subsets of patients with breast cancers that are HER2-RARα co-amplified and have low levels of MYC.


Asunto(s)
Neoplasias de la Mama , Resistencia a Antineoplásicos , Proteínas Proto-Oncogénicas c-myc , Receptor ErbB-2 , Receptores de Ácido Retinoico , Receptor alfa de Ácido Retinoico , Trastuzumab , Tretinoina , Humanos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Resistencia a Antineoplásicos/genética , Resistencia a Antineoplásicos/efectos de los fármacos , Receptor ErbB-2/metabolismo , Receptor ErbB-2/genética , Receptores de Ácido Retinoico/metabolismo , Receptores de Ácido Retinoico/genética , Femenino , Tretinoina/farmacología , Receptor alfa de Ácido Retinoico/genética , Receptor alfa de Ácido Retinoico/metabolismo , Trastuzumab/farmacología , Trastuzumab/uso terapéutico , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico
5.
Proc Natl Acad Sci U S A ; 121(18): e2317690121, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38648485

RESUMEN

The underlying mechanism(s) by which the PML::RARA fusion protein initiates acute promyelocytic leukemia is not yet clear. We defined the genomic binding sites of PML::RARA in primary mouse and human hematopoietic progenitor cells with V5-tagged PML::RARA, using anti-V5-PML::RARA chromatin immunoprecipitation sequencing and CUT&RUN approaches. Most genomic PML::RARA binding sites were found in regions that were already chromatin-accessible (defined by ATAC-seq) in unmanipulated, wild-type promyelocytes, suggesting that these regions are "open" prior to PML::RARA expression. We found that GATA binding motifs, and the direct binding of the chromatin "pioneering factor" GATA2, were significantly enriched near PML::RARA binding sites. Proximity labeling studies revealed that PML::RARA interacts with ~250 proteins in primary mouse hematopoietic cells; GATA2 and 33 others require PML::RARA binding to DNA for the interaction to occur, suggesting that binding to their cognate DNA target motifs may stabilize their interactions. In the absence of PML::RARA, Gata2 overexpression induces many of the same epigenetic and transcriptional changes as PML::RARA. These findings suggested that PML::RARA may indirectly initiate its transcriptional program by activating Gata2 expression: Indeed, we demonstrated that inactivation of Gata2 prior to PML::RARA expression prevented its ability to induce self-renewal. These data suggested that GATA2 binding creates accessible chromatin regions enriched for both GATA and Retinoic Acid Receptor Element motifs, where GATA2 and PML::RARA can potentially bind and interact with each other. In turn, PML::RARA binding to DNA promotes a feed-forward transcriptional program by positively regulating Gata2 expression. Gata2 may therefore be required for PML::RARA to establish its transcriptional program.


Asunto(s)
Factor de Transcripción GATA2 , Células Madre Hematopoyéticas , Proteínas de Fusión Oncogénica , Animales , Humanos , Ratones , Sitios de Unión , Autorrenovación de las Células , Cromatina/metabolismo , ADN/metabolismo , Factor de Transcripción GATA2/metabolismo , Factor de Transcripción GATA2/genética , Células Madre Hematopoyéticas/metabolismo , Leucemia Promielocítica Aguda/metabolismo , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/patología , Proteínas de Fusión Oncogénica/metabolismo , Proteínas de Fusión Oncogénica/genética , Proteína de la Leucemia Promielocítica/metabolismo , Proteína de la Leucemia Promielocítica/genética , Unión Proteica , Receptor alfa de Ácido Retinoico/metabolismo , Receptor alfa de Ácido Retinoico/genética
6.
Am J Physiol Cell Physiol ; 326(6): C1735-C1752, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38618701

RESUMEN

The potential neurotoxic effects of propofol, an extensively utilized anesthetic, underline the urgency to comprehend its influence on neuronal health. Insights into the role of the retinoic acid receptor-α, small nucleolar RNA host gene 1, and brain-derived neurotrophic factor (RARα-Snhg1-Bdnf) network can offer significant advancements in minimizing these effects. The study targets the exploration of the RARα and Snhg1 regulatory network's influence on Bdnf expression in the realm of propofol-induced neurotoxicity. Harnessing the Gene Expression Omnibus (GEO) database and utilizing JASPAR and RNA-Protein Interaction Prediction (RPISeq) database for projections, the study embarks on an in-depth analysis employing both in vitro and in vivo models. The findings draw a clear link between propofol-induced neurotoxicity and the amplification of RAR signaling pathways, impacting hippocampal development and apoptosis and leading to increased RARα and Snhg1 and decreased Bdnf. Propofol is inferred to accentuate neurotoxicity by heightening RARα and Snhg1 interactions, culminating in Bdnf suppression.NEW & NOTEWORTHY This study aimed to decode propofol's neurotoxic effects on the regulatory cascade, provide insights into the RARα-Snhg1-Bdnf interaction, apply extensive validation techniques, provide a detailed analysis and exploration of propofol's neurotoxicity, and offer a comprehensive approach to understanding molecular interactions.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo , Propofol , Receptor alfa de Ácido Retinoico , Propofol/toxicidad , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/genética , Receptor alfa de Ácido Retinoico/genética , Receptor alfa de Ácido Retinoico/metabolismo , Animales , Humanos , Transducción de Señal/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Síndromes de Neurotoxicidad/genética , Síndromes de Neurotoxicidad/metabolismo , Ratas , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Apoptosis/efectos de los fármacos , Masculino
7.
Proc Natl Acad Sci U S A ; 121(7): e2311803121, 2024 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-38330015

RESUMEN

Chronic kidney disease (CKD) is characterized by a gradual loss of kidney function and affects ~13.4% of the global population. Progressive tubulointerstitial fibrosis, driven in part by proximal tubule (PT) damage, is a hallmark of late stages of CKD and contributes to the development of kidney failure, for which there are limited treatment options. Normal kidney development requires signaling by vitamin A (retinol), which is metabolized to retinoic acid (RA), an endogenous agonist for the RA receptors (RARα, ß, γ). RARα levels are decreased in a mouse model of diabetic nephropathy and restored with RA administration; additionally, RA treatment reduced fibrosis. We developed a mouse model in which a spatiotemporal (tamoxifen-inducible) deletion of RARα in kidney PT cells of adult mice causes mitochondrial dysfunction, massive PT injury, and apoptosis without the use of additional nephrotoxic substances. Long-term effects (3 to 4.5 mo) of RARα deletion include increased PT secretion of transforming growth factor ß1, inflammation, interstitial fibrosis, and decreased kidney function, all of which are major features of human CKD. Therefore, RARα's actions in PTs are crucial for PT homeostasis, and loss of RARα causes injury and a key CKD phenotype.


Asunto(s)
Riñón , Insuficiencia Renal Crónica , Receptor alfa de Ácido Retinoico , Animales , Humanos , Ratones , Modelos Animales de Enfermedad , Fibrosis , Riñón/metabolismo , Túbulos Renales Proximales/metabolismo , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/prevención & control , Receptor alfa de Ácido Retinoico/genética , Receptor alfa de Ácido Retinoico/metabolismo , Tretinoina/farmacología , Tretinoina/metabolismo
8.
Am J Hematol ; 99(5): 1005-1007, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38410879

RESUMEN

IRF2BP1 breaked in the middle of exon 1 at the c.322 position and fused with RARA intron 2 which is located at 3717 bp upstream of its exon 3. The fusion produced a new intron by forming a paired splicing donor GT at 9 bp downstream of RARA breakpoint and acceptor AG at the 5' end of RARA exon 3. The IRF2BP1::RARA fusion gene leads a fusion transcript involving IRF2BP1 exon 1 and RARA exon 3, linked by a 9-bp fragment derived from RARA intron 2. The patient with IRF2BP1::RARA has same clinical features of APL.


Asunto(s)
Leucemia Promielocítica Aguda , Humanos , Cromosomas Humanos Par 17 , Exones/genética , Leucemia Promielocítica Aguda/genética , Proteínas de Fusión Oncogénica/genética , Receptores de Ácido Retinoico/genética , Receptor alfa de Ácido Retinoico/genética , Translocación Genética
9.
Ann Hematol ; 103(4): 1181-1185, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38294534

RESUMEN

Acute promyelocytic leukemia (APL) is a specific subtype of acute myeloid leukemia that is distinguished by the chromosomal translocation t(15;17)(q24;q21), which leads to the fusion of the promyelocytic leukemia (PML) gene with the retinoic acid receptor alpha (RARA). Recently, we identified a novel fusion gene in APL, RARA::ankyrin repeat domain 34C (ANKRD34C), identified its functions by morphological, cytogenetic, molecular biological and multiplex fluorescence in situ hybridization analyses, and demonstrated the potential therapeutic effect clinically and experimentally of all-trans retinoic acid (ATRA); the findings have important implications for the diagnosis and treatment of atypical APL.


Asunto(s)
Leucemia Promielocítica Aguda , Humanos , Leucemia Promielocítica Aguda/diagnóstico , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/tratamiento farmacológico , Hibridación Fluorescente in Situ , Tretinoina/uso terapéutico , Receptor alfa de Ácido Retinoico/genética , Proteínas Portadoras/genética , Translocación Genética , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo
10.
J Mol Endocrinol ; 72(1)2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-37930232

RESUMEN

There is increasing interest in retinoic acid (RA) as a regulator of the complex biological processes underlying the cognitive functions performed by the brain. The importance of RA in brain function is underlined by the brain's high efficiency in converting vitamin A into RA. One crucial action of RA in the brain is dependent on RA receptor α (RARα) transport out of the nucleus, where it no longer regulates transcription but carries out non-genomic functions. RARα, when localised in the cytoplasm, particularly in neuronal dendrites, acts as a translational suppressor. It regulates protein translation as a crucial part of the mechanism maintaining homoeostatic synaptic plasticity, which is characterised by neuronal changes necessary to restore and balance the excitability of neuronal networks after perturbation events. Under normal conditions of neurotransmission, RARα without ligand suppresses the translation of proteins. When neural activity is reduced, RA synthesis is stimulated, and RA signalling via RARα derepresses the translation of proteins and synergistically with the fragile X mental retardation protein allows the synthesis of Ca2+ permeable α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors that re-establish normal levels of synaptic activity. Homoeostatic synaptic plasticity underlies many cognitive processes, so its impairment due to dysregulation of RA signalling may be involved in neurodevelopmental disorders such as autism, which is also associated with FMRP. A full understanding of RA signalling control of homoeostatic synaptic plasticity may point to treatments.


Asunto(s)
Disfunción Cognitiva , Tretinoina , Humanos , Tretinoina/farmacología , Tretinoina/metabolismo , Receptores de Ácido Retinoico , Homeostasis/fisiología , Receptor alfa de Ácido Retinoico/genética , Plasticidad Neuronal
11.
Arch. endocrinol. metab. (Online) ; 62(3): 366-369, May-June 2018. graf
Artículo en Inglés | LILACS | ID: biblio-1038490

RESUMEN

ABSTRACT Objective: Graves' ophthalmopathy (GO) is an autoimmune disease that leads to ocular proptosis caused by fat accumulation and inflammation, and the main treatment is corticosteroid therapy. Retinoid acid receptor-alpha (RARα) seems to be associated with inflammation and adipocyte differentiation. This study aimed to assess the effect of glucocorticoid treatment on orbital fibroblasts of GO patient treated or not with different glucocorticoid doses. Materials and methods: Orbital fibroblasts collected during orbital decompression of a female patient with moderately severe/severe GO were cultivated and treated with 10 nM and 100 nM dexamethasone (Dex). rRARα gene expression in the treated and untreated cells was then compared. Results: Fibroblast RARα expression was not affected by 100 nM Dex. On the other hand, RARα expression was 24% lower in cells treated with 10 nM Dex (p < 0.05). Conclusions: Orbital fibroblasts from a GO patient expressed the RARα gene, which was unaffected by higher, but decreased with lower doses of glucocorticoid.


Asunto(s)
Humanos , Órbita/efectos de los fármacos , Dexametasona/administración & dosificación , Expresión Génica/efectos de los fármacos , Oftalmopatía de Graves/tratamiento farmacológico , Fibroblastos/química , Glucocorticoides/administración & dosificación , Órbita/patología , Índice de Severidad de la Enfermedad , Oftalmopatía de Graves/patología , Fibroblastos/efectos de los fármacos , Receptor alfa de Ácido Retinoico/efectos de los fármacos , Receptor alfa de Ácido Retinoico/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...