Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 121(9): e2320129121, 2024 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-38377195

RESUMEN

Despite numerous female contraceptive options, nearly half of all pregnancies are unintended. Family planning choices for men are currently limited to unreliable condoms and invasive vasectomies with questionable reversibility. Here, we report the development of an oral contraceptive approach based on transcriptional disruption of cyclical gene expression patterns during spermatogenesis. Spermatogenesis involves a continuous series of self-renewal and differentiation programs of spermatogonial stem cells (SSCs) that is regulated by retinoic acid (RA)-dependent activation of receptors (RARs), which control target gene expression through association with corepressor proteins. We have found that the interaction between RAR and the corepressor silencing mediator of retinoid and thyroid hormone receptors (SMRT) is essential for spermatogenesis. In a genetically engineered mouse model that negates SMRT-RAR binding (SMRTmRID mice), the synchronized, cyclic expression of RAR-dependent genes along the seminiferous tubules is disrupted. Notably, the presence of an RA-resistant SSC population that survives RAR de-repression suggests that the infertility attributed to the loss of SMRT-mediated repression is reversible. Supporting this notion, we show that inhibiting the action of the SMRT complex with chronic, low-dose oral administration of a histone deacetylase inhibitor reversibly blocks spermatogenesis and fertility without affecting libido. This demonstration validates pharmacologic targeting of the SMRT repressor complex for non-hormonal male contraception.


Asunto(s)
Proteínas de Unión al ADN , Proteínas Represoras , Humanos , Femenino , Masculino , Animales , Ratones , Proteínas de Unión al ADN/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Proteínas Co-Represoras/genética , Co-Represor 2 de Receptor Nuclear/genética , Tretinoina/farmacología , Anticoncepción , Co-Represor 1 de Receptor Nuclear
2.
J Biol Chem ; 299(10): 105255, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37714463

RESUMEN

9-cis-retinoic acid (9cRA) binds retinoic acid receptors (RAR) and retinoid X receptors (RXR) with nanomolar affinities, in contrast to all-trans-retinoic acid (atRA), which binds only RAR with nanomolar affinities. RXR heterodimerize with type II nuclear receptors, including RAR, to regulate a vast gene array. Despite much effort, 9cRA has not been identified as an endogenous retinoid, other than in pancreas. By revising tissue analysis methods, 9cRA quantification by liquid chromatography-tandem mass spectrometry becomes possible in all mouse tissues analyzed. 9cRA occurs in concentrations similar to or greater than atRA. Fasting increases 9cRA in white and brown adipose, brain and pancreas, while increasing atRA in white adipose, liver and pancreas. 9cRA supports FoxO1 actions in pancreas ß-cells and counteracts glucose actions that lead to glucotoxicity; in part by inducing Atg7 mRNA, which encodes the key enzyme essential for autophagy. Glucose suppresses 9cRA biosynthesis in the ß-cell lines 832/13 and MIN6. Glucose reduces 9cRA biosynthesis in 832/13 cells by inhibiting Rdh5 transcription, unconnected to insulin, through cAMP and Akt, and inhibiting FoxO1. Through adapting tissue specifically to fasting, 9cRA would act independent of atRA. Widespread occurrence of 9cRA in vivo, and its self-sufficient adaptation to energy status, provides new perspectives into regulation of energy balance, attenuation of insulin and glucose actions, regulation of type II nuclear receptors, and retinoid biology.


Asunto(s)
Alitretinoína , Metabolismo Energético , Glucosa , Células Secretoras de Insulina , Animales , Ratones , Alitretinoína/metabolismo , Glucosa/metabolismo , Glucosa/farmacología , Insulina/metabolismo , Tretinoina/metabolismo , Ratones Endogámicos C57BL , Ratas , Línea Celular , Regulación de la Expresión Génica/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Ayuno , Proteínas Proto-Oncogénicas c-akt/metabolismo
3.
J Biol Chem ; 299(5): 104669, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37011860

RESUMEN

Considerable evidence confirms the importance of Cyp26a1 to all-trans-retinoic acid (RA) homeostasis during embryogenesis. In contrast, despite its presence in postnatal liver as a potential major RA catabolizing enzyme and its acute sensitivity to induction by RA, some data suggested that Cyp26a1 contributes only marginally to endogenous RA homeostasis postnatally. We report reevaluation of a conditional Cyp26a1 knockdown in the postnatal mouse. The current results show that Cyp26a1 mRNA in WT mouse liver increases 16-fold upon refeeding after a fast, accompanied by an increased rate of RA elimination and a 41% decrease in the RA concentration. In contrast, Cyp26a1 mRNA in the refed homozygotic knockdown reached only 2% of its extent in WT during refeeding, accompanied by a slower rate of RA catabolism and no decrease in liver RA, relative to fasting. Refed homozygous knockdown mice also had decreased Akt1 and 2 phosphorylation and pyruvate dehydrogenase kinase 4 (Pdk4) mRNA and increased glucokinase (Gck) mRNA, glycogen phosphorylase (Pygl) phosphorylation, and serum glucose, relative to WT. Fasted homozygous knockdown mice had increased glucagon/insulin relative to WT. These data indicate that Cyp26a1 participates prominently in moderating the postnatal liver concentration of endogenous RA and contributes essentially to glucoregulatory control.


Asunto(s)
Glucemia , Homeostasis , Ácido Retinoico 4-Hidroxilasa , Tretinoina , Animales , Ratones , Hígado/enzimología , Hígado/metabolismo , Ácido Retinoico 4-Hidroxilasa/genética , Ácido Retinoico 4-Hidroxilasa/metabolismo , ARN Mensajero/genética , Tretinoina/metabolismo , Glucoquinasa/metabolismo , Glucógeno Fosforilasa/metabolismo , Insulina/metabolismo , Animales Recién Nacidos , Fosforilación , Glucemia/metabolismo
4.
Nutrients ; 14(21)2022 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-36364786

RESUMEN

All-trans-retinoic acid (RA), a metabolite of vitamin A (retinol), exerts profuse actions that enable multiple aspects of reproduction, embryonic development and post-natal regulation of energy metabolism, glucoregulatory control, organ function, and of the skeletal, immune, nervous and cardiovascular systems, as well as cell proliferation vs [...].


Asunto(s)
Tretinoina , Vitamina A , Embarazo , Femenino , Humanos , Tretinoina/metabolismo , Vitamina A/metabolismo , Autacoides
5.
iScience ; 25(7): 104564, 2022 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-35789854

RESUMEN

Retinoic acid (RA) counters insulin's metabolic actions. Insulin reduces liver RA biosynthesis by exporting FoxO1 from nuclei. RA induces its catabolism, catalyzed by CYP26A1. A CYP26A1 contribution to RA homeostasis with changes in energy status had not been investigated. We found that glucagon, cortisol, and dexamethasone decrease RA-induced CYP26A1 transcription, thereby reducing RA oxidation during fasting. Interaction between the glucocorticoid receptor and the RAR/RXR coactivation complex suppresses CYP26A1 expression, increasing RA's elimination half-life. Interaction between CCAAT-enhancer-binding protein beta (C/EBPß) and the major allele of SNP rs2068888 enhances CYP26A1 expression; the minor allele restricts the C/EBPß effect on CYP26A1. The major and minor alleles associate with impaired human health or reduction in blood triglycerides, respectively. Thus, regulating CYP26A1 transcription contributes to adapting RA to coordinate energy availability with metabolism. These results enhance insight into CYP26A1 effects on RA during changes in energy status and glucocorticoid receptor modification of RAR-regulated gene expression.

6.
Nutrients ; 14(8)2022 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-35458115

RESUMEN

This review addresses the fasting vs. re-feeding effects of retinoic acid (RA) biosynthesis and functions, and sexually dimorphic RA actions. It also discusses other understudied topics essential for understanding RA activities-especially interactions with energy-balance-regulating hormones, including insulin and glucagon, and sex hormones. This report will introduce RA homeostasis and hormesis to provide context. Essential context also will encompass RA effects on adiposity, muscle function and pancreatic islet development and maintenance. These comments provide background for explaining interactions among insulin, glucagon and cortisol with RA homeostasis and function. One aim would clarify the often apparent RA contradictions related to pancreagenesis vs. pancreas hormone functions. The discussion also will explore the adverse effects of RA on estrogen action, in contrast to the enhancing effects of estrogen on RA action, the adverse effects of androgens on RA receptors, and the RA induction of androgen biosynthesis.


Asunto(s)
Insulina , Tretinoina , Estrógenos/farmacología , Glucagón , Receptores de Ácido Retinoico , Tretinoina/farmacología
7.
J Biol Chem ; 297(3): 101101, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34419449

RESUMEN

The retinol dehydrogenase Rdh10 catalyzes the rate-limiting reaction that converts retinol into retinoic acid (RA), an autacoid that regulates energy balance and reduces adiposity. Skeletal muscle contributes to preventing adiposity, by consuming nearly half the energy of a typical human. We report sexually dimorphic differences in energy metabolism and muscle function in Rdh10+/- mice. Relative to wild-type (WT) controls, Rdh10+/- males fed a high-fat diet decrease reliance on fatty-acid oxidation and experience glucose intolerance and insulin resistance. Running endurance decreases 40%. Rdh10+/- females fed this diet increase fatty acid oxidation and experience neither glucose intolerance nor insulin resistance. Running endurance increases 220%. We therefore assessed RA function in the mixed-fiber type gastrocnemius muscles (GM), which contribute to running, rather than standing, and are similar to human GM. RA levels in Rdh10+/- male GM decrease 38% relative to WT. Rdh10+/- male GM increase expression of Myog and reduce Eif6 mRNAs, which reduce and enhance running endurance, respectively. Cox5A, complex IV activity, and ATP decrease. Increased centralized nuclei reveal existence of muscle malady and/or repair in GM fibers. Comparatively, RA in Rdh10+/- female GM decreases by less than half the male decrease, from a more modest decrease in Rdh10 and an increase in the estrogen-induced retinol dehydrogenase Dhrs9. Myog mRNA decreases. Cox5A, complex IV activity, and ATP increase. Centralized GM nuclei do not increase. We conclude that Rdh10/RA affects whole body energy use and insulin resistance partially through sexual dimorphic effects on skeletal muscle gene expression, structure, and mitochondria activity.


Asunto(s)
Oxidorreductasas de Alcohol/metabolismo , Músculo Esquelético/metabolismo , Adiposidad , Oxidorreductasas de Alcohol/genética , Animales , Dieta Alta en Grasa , Complejo IV de Transporte de Electrones/metabolismo , Metabolismo Energético/genética , Metabolismo Energético/fisiología , Femenino , Intolerancia a la Glucosa/metabolismo , Resistencia a la Insulina , Metabolismo de los Lípidos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/fisiología , Músculos/metabolismo , Oxidación-Reducción , Resistencia Física/fisiología , Carrera/fisiología , Caracteres Sexuales , Factores Sexuales , Tretinoina/metabolismo
8.
Methods Enzymol ; 637: 27-54, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32359649

RESUMEN

Generation of the autacoid all-trans-retinoic acid (ATRA) from retinol (vitamin A) relies on a complex metabolon that includes retinol binding-proteins and enzymes from the short-chain dehydrogenase/reductase and aldehyde dehydrogenase gene families. Serum retinol binding-protein delivers all-trans-retinol (vitamin A) from blood to cells through two membrane receptors, Stra6 and Rbpr2. Stra6 and Rbpr2 convey retinol to cellular retinol binding-protein type 1 (Crbp1). Holo-Crbp1 delivers retinol to lecithin: retinol acyl transferase (Lrat) for esterification and storage. Lrat channels retinol directly into its active site from holo-Crbp1 by protein-protein interaction. The ratio apo-Crbp1/holo-Crbp1 directs flux of retinol into and out of retinyl esters, through regulating esterification vs ester hydrolysis. Multiple retinol dehydrogenases (Rdh1, Rdh10, Dhrs9, Rdhe2, Rdhe2s) channel retinol from holo-Crbp1 to generate retinal for ATRA biosynthesis. ß-Carotene oxidase type 1 generates retinal from carotenoids, delivered by the scavenger receptor-B1. Retinal reductases (Dhrs3, Dhrs4, Rdh11) reduce retinal into retinol, thereby restraining ATRA biosynthesis. Retinal dehydrogenases (Raldh1, 2, 3) dehydrogenate retinal irreversibly into ATRA. ATRA regulates its own concentrations by inducing Lrat and ATRA degradative enzymes. ATRA exhibits hormesis. Its effects relate to its concentration as an inverted J-shaped curve, transitioning from beneficial in the "goldilocks" zone to toxicity, as concentrations increase. Hormesis has distorted understanding physiological effects of ATRA post-nataly using chow-diet fed, ATRA-dosed animal models. Cancer, immune deficiency and metabolic abnormalities result from mutations and/or insufficiency in Crbp1 and retinoid metabolizing enzymes.


Asunto(s)
Tretinoina , Vitamina A , Animales , Proteínas de Unión al Retinol , Proteínas Celulares de Unión al Retinol/genética , Tretinoina/metabolismo , Vitamina A/metabolismo
9.
Methods Enzymol ; 637: 55-75, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32359659

RESUMEN

Cellular retinoid-binding proteins (BP) chaperone retinol through esterification, conversion of retinol into retinal, reduction of retinal, conversion of retinal into all-trans-retinoic acid (ATRA), and ATRA to catabolism. They also deliver ATRA to nuclear receptors and mediate non-genomic ATRA actions. These retinoid-specific binding-proteins include: cellular retinol binding-protein, type 1 (Crbp1), cellular retinol binding-protein type 2 (Crbp2), cellular retinol binding-protein type 3 (Crbp3), cellular retinoic acid binding-protein type 1 (Crabp1); cellular retinoic acid binding-protein type 2 (Crabp2). Retinoid BP bind their ligands specifically and with high-affinity. These BP seemingly evolved to solubilize the lipophilic retinoids in the aqueous cellular medium, and allow retinoid access only to enzymes that recognize both the BP and the retinoid. By chaperoning retinoids through cells, retinoid BP provide specificity to retinoids' metabolism and protect the scarce resource from dispersing into cell membranes and/or undergoing catabolism as xenobiotics. Other functions include non-genomic actions of Crabp1, delivery of ATRA to RAR by holo-Crabp2, and stabilization of HuR by apo-Crabp2. In addition to the retinoid-specific BP, Fabp5 also binds ATRA and delivers it to Pparδ. This article describes these BP and their functions, with a focus on experimental protocols to distinguish protein-protein interactions from diffusion-mediated transfer of ligand from BP to enzymes or receptors, and methods for quantifying retinoids.


Asunto(s)
Retinoides , Proteínas de Unión al Retinol , Retinoides/metabolismo , Proteínas Celulares de Unión al Retinol , Tretinoina/metabolismo , Vitamina A/metabolismo
10.
Anal Chem ; 91(22): 14624-14630, 2019 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-31644264

RESUMEN

We report a high-performance, liquid chromatography/tandem mass spectrometry (HPLC-MS/MS) assay to quantify without derivatizaton dehyroepiandrosterone (DHEA), 17ß-estradiol (E2), testosterone (T), and their sulfates in serum and tissues. This assay functions well with multiple adipose depots, a previously unattained analysis. To delipidate and facilitate recovery, tissues were homogenized in acetonitrile, and the homogenate was frozen. The supernatant was evaporated, resuspended in an aqueous acetate buffer, and extracted with hexane to separate free (unconjugated) from sulfated steroids. Sulfated steroids in the aqueous medium were then hydrolyzed with sulfatase and extracted with hexane. Each extract was analyzed separately. HPLC resolution combined with the sensitivity and specificity of MS/MS allowed quantification of DHEA, E2, and T with 10, 10, and 5 fmol lower limits of quantification and linear ranges to 1 pmol. Application of the method to mouse serum and tissues reveals ranges of DHEA, E2, and T and their sulfates, and tissue-specific differences in steroid profile, especially white versus brown adipose. In addition, marginal decreases of T in all tissues and considerable increases in DHEA in male iWAT and eWAT in response to a high-fat diet further strengthen the inference regarding the role of steroid metabolism in adipogenesis. This assay permits detailed studies of interactions between adiposity and sex steroids in serum and tissues, including adipose.


Asunto(s)
Deshidroepiandrosterona/sangre , Estradiol/sangre , Ésteres del Ácido Sulfúrico/sangre , Testosterona/sangre , Animales , Cromatografía Liquida/métodos , Femenino , Límite de Detección , Extracción Líquido-Líquido , Masculino , Ratones Endogámicos C57BL , Espectrometría de Masas en Tándem/métodos
11.
Proc Natl Acad Sci U S A ; 116(8): 3126-3135, 2019 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-30718413

RESUMEN

The balance of effector versus regulatory T cells (Tregs) controls inflammation in numerous settings, including multiple sclerosis (MS). Here we show that memory phenotype CD4+ T cells infiltrating the central nervous system during experimental autoimmune encephalomyelitis (EAE), a widely studied animal model of MS, expressed high levels of mRNA for Dgat1 encoding diacylglycerol-O-acyltransferase-1 (DGAT1), an enzyme that catalyzes triglyceride synthesis and retinyl ester formation. DGAT1 inhibition or deficiency attenuated EAE, with associated enhanced Treg frequency; and encephalitogenic, DGAT1-/- in vitro-polarized Th17 cells were poor inducers of EAE in adoptive recipients. DGAT1 acyltransferase activity sequesters retinol in ester form, preventing synthesis of retinoic acid, a cofactor for Treg generation. In cultures with T cell-depleted lymphoid tissues, retinol enhanced Treg induction from DGAT1-/- but not from WT T cells. The WT Treg induction defect was reversed by DGAT1 inhibition. These results demonstrate that DGAT1 suppresses retinol-dependent Treg formation and suggest its potential as a therapeutic target for autoimmune inflammation.


Asunto(s)
Diacilglicerol O-Acetiltransferasa/genética , Encefalomielitis/genética , Inflamación/genética , Esclerosis Múltiple/genética , Linfocitos T Reguladores/inmunología , Animales , Sistema Nervioso Central , Técnicas de Inactivación de Genes , Humanos , Inflamación/inmunología , Inflamación/patología , Ratones , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Células TH1/inmunología , Células Th17/inmunología , Tretinoina/metabolismo
12.
Cell Mol Life Sci ; 76(12): 2425-2447, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30788515

RESUMEN

RDH1 is one of the several enzymes that catalyze the first of the two reactions to convert retinol into all-trans-retinoic acid (atRA). Here, we show that Rdh1-null mice fed a low-fat diet gain more weight as adiposity (17% males, 13% females) than wild-type mice by 20 weeks old, despite neither consuming more calories nor decreasing activity. Glucose intolerance and insulin resistance develop following increased adiposity. Despite the increase in white fat pads, epididymal white adipose does not express Rdh1, nor does muscle. Brown adipose tissue (BAT) and liver express Rdh1 at relatively high levels compared to other tissues. Rdh1 ablation lowered body temperatures during ambient conditions. Given the decreased body temperature, we focused on BAT. A lack of differences in BAT adipogenic gene expression between Rdh1-null mice and wild-type mice, including Pparg, Prdm16, Zfp516 and Zfp521, indicated that the phenotype was not driven by brown adipose hyperplasia. Rather, Rdh1 ablation eliminated the increase in BAT atRA that occurs after re-feeding. This disruption of atRA homeostasis increased fatty acid uptake, but attenuated lipolysis in primary brown adipocytes, resulting in increased lipid content and larger lipid droplets. Rdh1 ablation also decreased mitochondrial proteins, including CYCS and UCP1, the mitochondria oxygen consumption rate, and disrupted the mitochondria membrane potential, further reflecting impaired BAT function, resulting in both BAT and white adipose hypertrophy. RNAseq revealed dysregulation of 424 BAT genes in null mice, which segregated predominantly into differences after fasting vs after re-feeding. Exceptions were Rbp4 and Gbp2b, which increased during both dietary conditions. Rbp4 encodes the serum retinol-binding protein-an insulin desensitizer. Gbp2b encodes a GTPase. Because Gbp2b increased several hundred-fold, we overexpressed it in brown adipocytes. This caused a shift to larger lipid droplets, suggesting that GBP2b affects signaling downstream of the ß-adrenergic receptor during basal thermogenesis. Thus, Rdh1-generated atRA in BAT regulates multiple genes that promote BAT adaptation to whole-body energy status, such as fasting and re-feeding. These gene expression changes promote optimum mitochondria function and thermogenesis, limiting adiposity. Attenuation of adiposity and insulin resistance suggests that RDH1 mitigates metabolic syndrome.


Asunto(s)
Tejido Adiposo Pardo/fisiología , Adiposidad , Ayuno , Hidroxiesteroide Deshidrogenasas/metabolismo , Tretinoina/metabolismo , Animales , Dieta con Restricción de Grasas , Ingestión de Alimentos , Metabolismo Energético , Femenino , Eliminación de Gen , Intolerancia a la Glucosa/genética , Intolerancia a la Glucosa/metabolismo , Hidroxiesteroide Deshidrogenasas/genética , Resistencia a la Insulina , Metabolismo de los Lípidos , Masculino , Ratones Endogámicos C57BL , Termogénesis , Vitamina A/metabolismo
13.
Artículo en Inglés | MEDLINE | ID: mdl-30455235

RESUMEN

Fluconazole-induced alopecia is a significant problem for patients receiving long-term therapy. We evaluated the hair cycle changes of fluconazole in a rat model and investigated potential molecular mechanisms. Plasma and tissue levels of retinoic acid were not found to be causal. Human patients with alopecia attributed to fluconazole also underwent detailed assessment and in both our murine model and human cohort fluconazole induced telogen effluvium. Future work further examining the mechanism of fluconazole-induced alopecia should be undertaken.


Asunto(s)
Alopecia Areata/inducido químicamente , Antifúngicos/efectos adversos , Fluconazol/efectos adversos , Alopecia Areata/sangre , Alopecia Areata/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Ratas , Ratas Wistar , Tretinoina/sangre , Tretinoina/metabolismo
14.
Stem Cells ; 36(9): 1368-1379, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29781215

RESUMEN

Retinoic acid (RA) signaling is essential for the differentiation of embryonic stem cells (ESCs) and vertebrate development. RA biosynthesis and metabolism are controlled by a series of enzymes, but the molecular regulators of these enzymes remain largely obscure. In this study, we investigated the functional role of the WD-domain protein STRAP (serine threonine kinase receptor-associated protein) in the pluripotency and lineage commitment of murine ESCs. We generated Strap knockout (KO) mouse ESCs and subjected them to spontaneous differentiation. We observed that, despite the unchanged characteristics of ESCs, Strap KO ESCs exhibited defects for lineage differentiation. Signature gene expression analyses revealed that Strap deletion attenuated intracellular RA signaling in embryoid bodies (EBs), and exogenous RA significantly rescued this deficiency. Moreover, loss of Strap selectively induced Cyp26A1 expression in mouse EBs, suggesting a potential role of STRAP in RA signaling. Mechanistically, we identified putative Krüppel-like factor 9 (KLF9) binding motifs to be critical in the enhancement of non-canonical RA-induced transactivation of Cyp26A1. Increased KLF9 expression in the absence of STRAP is partially responsible for Cyp26A1 induction. Interestingly, STRAP knockdown in Xenopus embryos influenced anterior-posterior neural patterning and impaired the body axis and eye development during early Xenopus embryogenesis. Taken together, our study reveals an intrinsic role for STRAP in the regulation of RA signaling and provides new molecular insights for ESC fate determination. Stem Cells 2018;36:1368-1379.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/deficiencia , Células Madre Embrionarias de Ratones/metabolismo , Ácido Retinoico 4-Hidroxilasa/metabolismo , Tretinoina/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Diferenciación Celular/fisiología , Linaje de la Célula , Células Cultivadas , Homeostasis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/enzimología , Ácido Retinoico 4-Hidroxilasa/genética , Transducción de Señal , Xenopus laevis
15.
Diabetes ; 67(4): 662-673, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29321172

RESUMEN

Pharmacological dosing of all-trans-retinoic acid (atRA) controls adiposity in rodents by inhibiting adipogenesis and inducing fatty acid oxidation. Retinol dehydrogenases (Rdh) catalyze the first reaction that activates retinol into atRA. This study examined postnatal contributions of Rdh10 to atRA biosynthesis and physiological functions of endogenous atRA. Embryonic fibroblasts from Rdh10 heterozygote hypomorphs or with a total Rdh10 knockout exhibit decreased atRA biosynthesis and escalated adipogenesis. atRA or a retinoic acid receptor (RAR) pan-agonist reversed the phenotype. Eliminating one Rdh10 copy in vivo (Rdh10+/- ) yielded a modest decrease (≤25%) in the atRA concentration of liver and adipose but increased adiposity in male and female mice fed a high-fat diet (HFD); increased liver steatosis, glucose intolerance, and insulin resistance in males fed an HFD; and activated bone marrow adipocyte formation in females, regardless of dietary fat. Chronic dosing with low-dose atRA corrected the metabolic defects. These data resolve physiological actions of endogenous atRA, reveal sex-specific effects of atRA in vivo, and establish the importance of Rdh10 to metabolic control by atRA. The consequences of a modest decrease in tissue atRA suggest that impaired retinol activation may contribute to diabesity, and low-dose atRA therapy may ameliorate adiposity and its sequelae of glucose intolerance and insulin resistance.


Asunto(s)
Adipogénesis/genética , Tejido Adiposo/metabolismo , Oxidorreductasas de Alcohol/genética , Metabolismo de los Lípidos/genética , Hígado/metabolismo , Tretinoina/metabolismo , Adipogénesis/efectos de los fármacos , Adiposidad/genética , Animales , Dieta Alta en Grasa , Femenino , Fibroblastos/metabolismo , Intolerancia a la Glucosa/metabolismo , Heterocigoto , Resistencia a la Insulina/genética , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Ratones , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Oxidación-Reducción , Receptores de Ácido Retinoico/agonistas , Factores Sexuales , Tretinoina/farmacología , Vitamina A/metabolismo
16.
PLoS One ; 12(11): e0187669, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29095919

RESUMEN

All-trans-retinoic acid (RA) inhibits adipogenesis in established preadipocyte cell lines. Dosing pharmacological amounts of RA reduces weight gain in mice fed a high-fat diet, i.e. counteracts diet-induced obesity (DIO). The aldehyde dehydrogenase Raldh1 (Aldh1a1) functions as one of three enzymes that converts the retinol metabolite retinal into RA, and one of many proteins that contribute to RA homeostasis. Female Raldh1-ablated mice resist DIO. This phenotype contrasts with ablations of other enzymes and binding-proteins that maintain RA homeostasis, which gain adiposity. The phenotype observed prompted the conclusion that loss of Raldh1 causes an increase in adipose tissue retinal, and therefore, retinal functions independently of RA to prevent DIO. A second deduction proposed that low nM concentrations of RA stimulate adipogenesis, in contrast to higher concentrations. Using peer-reviewed LC/MS/MS assays developed and validated for quantifying tissue RA and retinal, we show that endogenous retinal and RA concentrations in adipose tissues from Raldh1-null mice do not correlate with the phenotype. Moreover, male Raldh1-null mice resist weight gain regardless of dietary fat content. Resistance to weight gain occurs during adolescence in both sexes. We show that RA concentrations as low as 1 nM, i.e. in the sub-physiological range, impair adipogenesis of embryonic fibroblasts from wild-type mice. Embryonic fibroblasts from Raldh1-null mice resist differentiating into adipocytes, but retain ability to generate RA. These fibroblasts remain sensitive to an RA receptor pan-agonist, and are not affected by an RA receptor pan-antagonist. Thus, the data do not support the hypothesis that retinal itself represses weight gain and adipogenesis independently of RA. Instead, the data indicate that Raldh1 functions as a retinal and atRA-independent promoter of adiposity during adolescence, and enhances adiposity through pre-adipocyte cell autonomous actions.


Asunto(s)
Adiposidad , Isoenzimas/fisiología , Retinal-Deshidrogenasa/fisiología , Retinaldehído/metabolismo , Transducción de Señal , Familia de Aldehído Deshidrogenasa 1 , Animales , Ratones
17.
Pharmacol Ther ; 173: 19-33, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28132904

RESUMEN

Cellular binding-proteins (BP), including CRBP1, CRBP2, CRABP1, CRABP2, and FABP5, shepherd the poorly aqueous soluble retinoids during uptake, metabolism and function. Holo-BP promote efficient use of retinol, a scarce but essential nutrient throughout evolution, by sheltering it and its major metabolite all-trans-retinoic acid from adventitious interactions with the cellular milieu, and by imposing specificity of delivery to enzymes, nuclear receptors and other partners. Apo-BP reflect cellular retinoid status and modify activities of retinoid metabolon enzymes, or exert non-canonical actions. High ligand binding affinities and the nature of ligand sequestration necessitate external factors to prompt retinoid release from holo-BP. One or more of cross-linking, kinetics, and colocalization have identified these factors as RDH, RALDH, CYP26, LRAT, RAR and PPARß/δ. Michaelis-Menten and other kinetic approaches verify that BP channel retinoids to select enzymes and receptors by protein-protein interactions. Function of the BP and enzymes that constitute the retinoid metabolon depends in part on retinoid exchanges unique to specific pairings. The complexity of these exchanges configure retinol metabolism to meet the diverse functions of all-trans-retinoic acid and its ability to foster contrary outcomes in different cell types, such as inducing apoptosis, differentiation or proliferation. Altered BP expression affects retinoid function, for example, by impairing pancreas development resulting in abnormal glucose and energy metabolism, promoting predisposition to breast cancer, and fostering more severe outcomes in prostate cancer, ovarian adenocarcinoma, and glioblastoma. Yet, the extent of BP interactions with retinoid metabolon enzymes and their impact on retinoid physiology remains incompletely understood.


Asunto(s)
Retinoides/metabolismo , Proteínas Celulares de Unión al Retinol/metabolismo , Tretinoina/metabolismo , Animales , Proteínas de Unión a Ácidos Grasos/genética , Proteínas de Unión a Ácidos Grasos/metabolismo , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Humanos , Neoplasias/genética , Neoplasias/patología , Receptores de Ácido Retinoico/genética , Receptores de Ácido Retinoico/metabolismo , Proteínas Celulares de Unión al Retinol/genética
18.
Am J Respir Cell Mol Biol ; 56(2): 233-241, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27654457

RESUMEN

Immune-modulating drugs that target myeloid-derived suppressor cells or stimulate natural killer T cells have been shown to reduce mycobacterial loads in tuberculosis (TB). We aimed to determine if a combination of these drugs as adjunct immunotherapy to conventional antibiotic treatment could also increase therapeutic efficacy against TB. In our model of pulmonary TB in mice, we applied treatment with isoniazid, rifampicin, and pyrazinamide for 13 weeks alone or combined with immunotherapy consisting of all-trans retinoic acid, 1,25(OH)2-vitamin D3, and α-galactosylceramide. Outcome parameters were mycobacterial load during treatment (therapeutic activity) and 13 weeks after termination of treatment (therapeutic efficacy). Moreover, cellular changes were analyzed using flow cytometry and cytokine expression was assessed at the mRNA and protein levels. Addition of immunotherapy was associated with lower mycobacterial loads after 5 weeks of treatment and significantly reduced relapse of disease after a shortened 13-week treatment course compared with antibiotic treatment alone. This was accompanied by reduced accumulation of immature myeloid cells in the lungs at the end of treatment and increased TNF-α protein levels throughout the treatment period. We demonstrate, in a mouse model of pulmonary TB, that immunotherapy consisting of three clinically approved drugs can improve the therapeutic efficacy of standard antibiotic treatment.


Asunto(s)
Antibacterianos/uso terapéutico , Inmunoterapia , Tuberculosis/inmunología , Tuberculosis/terapia , Animales , Antibacterianos/farmacología , Antituberculosos/farmacología , Antituberculosos/uso terapéutico , Colecalciferol/farmacología , Colecalciferol/uso terapéutico , Terapia Combinada , Modelos Animales de Enfermedad , Femenino , Galactosilceramidas/farmacología , Galactosilceramidas/uso terapéutico , Inmunidad Celular/efectos de los fármacos , Pulmón/microbiología , Pulmón/patología , Ratones Endogámicos BALB C , Recurrencia , Tretinoina/sangre , Tuberculosis/sangre , Tuberculosis/tratamiento farmacológico , Factor de Necrosis Tumoral alfa/metabolismo
19.
Subcell Biochem ; 81: 21-76, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27830500

RESUMEN

Multiple binding and transport proteins facilitate many aspects of retinoid biology through effects on retinoid transport, cellular uptake, metabolism, and nuclear delivery. These include the serum retinol binding protein sRBP (aka Rbp4), the plasma membrane sRBP receptor Stra6, and the intracellular retinoid binding-proteins such as cellular retinol-binding proteins (CRBP) and cellular retinoic acid binding-proteins (CRABP). sRBP transports the highly lipophilic retinol through an aqueous medium. The major intracellular retinol-binding protein, CRBP1, likely enhances efficient retinoid use by providing a sink to facilitate retinol uptake from sRBP through the plasma membrane or via Stra6, delivering retinol or retinal to select enzymes that generate retinyl esters or retinoic acid, and protecting retinol/retinal from excess catabolism or opportunistic metabolism. Intracellular retinoic acid binding-proteins (CRABP1 and 2, and FABP5) seem to have more diverse functions distinctive to each, such as directing retinoic acid to catabolism, delivering retinoic acid to specific nuclear receptors, and generating non-canonical actions. Gene ablation of intracellular retinoid binding-proteins does not cause embryonic lethality or gross morphological defects. Metabolic and functional defects manifested in knockouts of CRBP1, CRBP2 and CRBP3, however, illustrate their essentiality to health, and in the case of CRBP2, to survival during limited dietary vitamin A. Future studies should continue to address the specific molecular interactions that occur between retinoid binding-proteins and their targets and their precise physiologic contributions to retinoid homeostasis and function.


Asunto(s)
Retinoides/fisiología , Proteínas Celulares de Unión al Retinol/fisiología , Oxidorreductasas de Alcohol/metabolismo , Aldehído Deshidrogenasa/metabolismo , Animales , Transporte Biológico , Núcleo Celular/metabolismo , Ojo/metabolismo , Técnicas de Inactivación de Genes , Homeostasis , Humanos , Mucosa Intestinal/metabolismo , Ratones , Ratones Noqueados , Modelos Moleculares , Proteínas de Neoplasias/metabolismo , Conformación Proteica , Receptores Citoplasmáticos y Nucleares/metabolismo , Retinaldehído/metabolismo , Proteínas Celulares de Unión al Retinol/química , Proteínas Celulares de Unión al Retinol/deficiencia , Proteínas Celulares de Unión al Retinol/genética , Transducción de Señal/fisiología , Tretinoina/metabolismo , Vitamina A/metabolismo , Vitamina A/toxicidad
20.
Cancer Immunol Res ; 4(11): 917-926, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27638841

RESUMEN

Chronic intestinal inflammation accompanies familial adenomatous polyposis (FAP) and is a major risk factor for colorectal cancer in patients with this disease, but the cause of such inflammation is unknown. Because retinoic acid (RA) plays a critical role in maintaining immune homeostasis in the intestine, we hypothesized that altered RA metabolism contributes to inflammation and tumorigenesis in FAP. To assess this hypothesis, we analyzed RA metabolism in the intestines of patients with FAP as well as APCMin/+ mice, a model that recapitulates FAP in most respects. We also investigated the impact of intestinal RA repletion and depletion on tumorigenesis and inflammation in APCMin/+ mice. Tumors from both FAP patients and APCMin/+ mice displayed striking alterations in RA metabolism that resulted in reduced intestinal RA. APCMin/+ mice placed on a vitamin A-deficient diet exhibited further reductions in intestinal RA with concomitant increases in inflammation and tumor burden. Conversely, restoration of RA by pharmacologic blockade of the RA-catabolizing enzyme CYP26A1 attenuated inflammation and diminished tumor burden. To investigate the effect of RA deficiency on the gut immune system, we studied lamina propria dendritic cells (LPDC) because these cells play a central role in promoting tolerance. APCMin/+ LPDCs preferentially induced Th17 cells, but reverted to inducing Tregs following restoration of intestinal RA in vivo or direct treatment of LPDCs with RA in vitro These findings demonstrate the importance of intestinal RA deficiency in tumorigenesis and suggest that pharmacologic repletion of RA could reduce tumorigenesis in FAP patients. Cancer Immunol Res; 4(11); 917-26. ©2016 AACR.


Asunto(s)
Antineoplásicos/farmacología , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/genética , Enterocolitis/genética , Genes APC , Tretinoina/farmacología , Adenoma/genética , Adenoma/metabolismo , Adenoma/patología , Poliposis Adenomatosa del Colon/complicaciones , Poliposis Adenomatosa del Colon/genética , Poliposis Adenomatosa del Colon/metabolismo , Poliposis Adenomatosa del Colon/patología , Animales , Transformación Celular Neoplásica/metabolismo , Neoplasias Colorrectales/etiología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Enterocolitis/tratamiento farmacológico , Enterocolitis/metabolismo , Enterocolitis/patología , Humanos , Ratones , Fenotipo , Células Th17/inmunología , Células Th17/metabolismo , Tretinoina/metabolismo , Carga Tumoral , Vitamina A/metabolismo , Deficiencia de Vitamina A/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA