Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Lupus ; 33(7): 716-727, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38616341

RESUMEN

OBJECTIVES: We aim to independently assess the validity of the damage index for antiphospholipid syndrome (DIAPS) in thrombotic antiphospholipid syndrome (APS) patients by exploring the prevalence and risk factors of organ damage and evaluating its impact on health-related quality of life (HR-QoL). METHODS: Cross-sectional study including all thrombotic APS patients (Sydney criteria) attending a Portuguese tertiary centre. Damage was assessed using the DIAPS, and HR-QoL using the 3- and 5-level EuroQol HR-QoL (EQ-D5-3L and 5L), and Visual Analogue Scale (VAS) applied via a phone questionnaire. Spearman's correlation between DIAPS and the HR-QoL scales was performed. Risk factors for damage accrual and HR-QoL impairment were explored using univariate and multivariate logistic regression. RESULTS: Among the 108 patients (female, 65.7%; white, 90.7%; primary APS, 75.9%; median disease duration, 6 years), damage (DIAPS≥1) developed in 48.2% of patients (mean ± SD DIAPS, 3.08 ± 1.83). DIAPS's neuropsychiatric domain was the most affected (24.2%), followed by the peripheral vascular domain (20.3%). No clinical, demographic nor laboratory parameters were significantly associated with damage. Regarding HR-QoL, pain/discomfort, anxiety/depression and usual activities domains were the most frequently impaired in both scales. DIAPS's domains correlated similarly with the EQ-5D-3L and 5L scales' individual domains. Female sex, medical disorders, secondary APS and type of presenting thrombosis (arterial) increased the risk of HR-QoL impairment. Total DIAPS was associated with higher odds of mobility, self-care and pain/discomfort impairment in both EQ-5D-3L and 5L scales but lost its independent risk in multivariable analysis. CONCLUSION: This external validation of DIAPS reinforces the ability of the score to correlate with HR-QoL while also highlighting risk factors for HR-QoL impairment other than damage accrual.


Asunto(s)
Síndrome Antifosfolípido , Calidad de Vida , Trombosis , Humanos , Síndrome Antifosfolípido/complicaciones , Femenino , Masculino , Adulto , Estudios Transversales , Persona de Mediana Edad , Factores de Riesgo , Trombosis/etiología , Encuestas y Cuestionarios , Portugal/epidemiología , Índice de Severidad de la Enfermedad , Modelos Logísticos
2.
J Am Soc Nephrol ; 32(2): 342-356, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33478973

RESUMEN

BACKGROUND: The kidney plays an important role in maintaining normal blood pH. Metabolic acidosis (MA) upregulates the pathway that mitochondria in the proximal tubule (PT) use to produce ammonia and bicarbonate from glutamine, and is associated with AKI. However, the extent to which MA causes AKI, and thus whether treating MA would be beneficial, is unclear. METHODS: Gavage with ammonium chloride induced acute MA. Multiphoton imaging of mitochondria (NADH/membrane potential) and transport function (dextran/albumin uptake), oxygen consumption rate (OCR) measurements in isolated tubules, histologic analysis, and electron microscopy in fixed tissue, and urinary biomarkers (KIM-1/clara cell 16) assessed tubular cell structure and function in mouse kidney cortex. RESULTS: MA induces an acute change in NAD redox state (toward oxidation) in PT mitochondria, without changing the mitochondrial energization state. This change is associated with a switch toward complex I activity and decreased maximal OCR, and a major alteration in normal lipid metabolism, resulting in marked lipid accumulation in PTs and the formation of large multilamellar bodies. These changes, in turn, lead to acute tubular damage and a severe defect in solute uptake. Increasing blood pH with intravenous bicarbonate substantially improves tubular function, whereas preinjection with the NAD precursor nicotinamide (NAM) is highly protective. CONCLUSIONS: MA induces AKI via changes in PT NAD and lipid metabolism, which can be reversed or prevented by treatment strategies that are viable in humans. These findings might also help to explain why MA accelerates decline in function in CKD.


Asunto(s)
Acidosis/etiología , Lesión Renal Aguda/etiología , Túbulos Renales/metabolismo , Túbulos Renales/patología , Metabolismo de los Lípidos/fisiología , NAD/metabolismo , Acidosis/metabolismo , Acidosis/patología , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Animales , Modelos Animales de Enfermedad , Corteza Renal/metabolismo , Corteza Renal/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Mitocondrias/patología , Consumo de Oxígeno/fisiología
3.
Kidney Int ; 100(3): 527-535, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34015315

RESUMEN

The development of intravital imaging with multiphoton microscopy has had a major impact on kidney research. It provides the unique opportunity to visualize dynamic behavior of cells and organelles in their native environment and to relate this to the complex 3-dimensional structure of the organ. Moreover, changes in cell/organelle function can be followed in real time in response to physiological interventions or disease-causing insults. However, realizing the enormous potential of this exciting approach has necessitated overcoming several substantial practical hurdles. In this article, we outline the nature of these challenges and how a variety of technical advances have provided effective solutions. In particular, improvements in laser/microscope technology, fluorescent probes, transgenic animals, and abdominal windows are collectively making previously opaque processes visible. Meanwhile, the rise of machine learning-based image analysis is facilitating the rapid generation of large amounts of quantitative data, amenable to deeper statistical interrogation. Taken together, the increased capabilities of multiphoton imaging are opening up huge new possibilities to study structure-function relationships in the kidney in unprecedented detail. In addition, they are yielding important new insights into cellular mechanisms of tissue damage, repair, and adaptive remodeling during disease states. Thus, intravital microscopy is truly entering an exciting new era in translational kidney research.


Asunto(s)
Microscopía Intravital , Microscopía de Fluorescencia por Excitación Multifotónica , Abdomen , Animales , Colorantes Fluorescentes , Riñón/diagnóstico por imagen
4.
FASEB J ; 34(6): 8510-8525, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32367531

RESUMEN

Alpha intercalated cells (αICs) in the kidney collecting duct (CD) belong to a family of mitochondria rich cells (MRCs) and have a crucial role in acidifying the urine via apical V-ATPase pumps. The nature of metabolism in αICs and its relationship to transport was not well-understood. Here, using multiphoton live cell imaging in mouse kidney tissue, FIB-SEM, and other complementary techniques, we provide new insights into mitochondrial structure and function in αICs. We show that αIC mitochondria have a rounded structure and are not located in close proximity to V-ATPase containing vesicles. They display a bright NAD(P)H fluorescence signal and low uptake of voltage-dependent dyes, but are energized by a pH gradient. However, expression of complex V (ATP synthase) is relatively low in αICs, even when stimulated by metabolic acidosis. In contrast, anaerobic glycolytic capacity is surprisingly high, and sufficient to maintain intracellular calcium homeostasis in the presence of complete aerobic inhibition. Moreover, glycolysis is essential for V-ATPase-mediated proton pumping. Key findings were replicated in narrow/clear cells in the epididymis, also part of the MRC family. In summary, using a range of cutting-edge techniques to investigate αIC metabolism in situ, we have discovered that these mitochondria dense cells have a high glycolytic capacity.


Asunto(s)
Glucólisis/fisiología , Túbulos Renales Colectores/metabolismo , Mitocondrias/metabolismo , Adenosina Trifosfatasas/metabolismo , Animales , Calcio/metabolismo , Epidídimo/metabolismo , Células Epiteliales/metabolismo , Homeostasis/fisiología , Concentración de Iones de Hidrógeno , Masculino , Ratones , Ratones Endogámicos C57BL , Bombas de Protones/metabolismo , ATPasas de Translocación de Protón/metabolismo
5.
Am J Physiol Renal Physiol ; 319(2): F245-F255, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32567348

RESUMEN

Ca2+ is an important second messenger that translates extracellular stimuli into intracellular responses. Although there has been significant progress in understanding Ca2+ dynamics in organs such as the brain, the nature of Ca2+ signals in the kidney is still poorly understood. Here, we show that by using a genetically expressed highly sensitive reporter (GCaMP6s), it is possible to perform imaging of Ca2+ signals at high resolution in the mouse kidney in vivo. Moreover, by applying machine learning-based automated analysis using a Ca2+-independent signal, quantitative data can be extracted in an unbiased manner. By projecting the resulting data onto the structure of the kidney, we show that different tubular segments display highly distinct spatiotemporal patterns of Ca2+ signals. Furthermore, we provide evidence that Ca2+ activity in the proximal tubule decreases with increasing distance from the glomerulus. Finally, we demonstrate that substantial changes in intracellular Ca2+ can be detected in proximal tubules in a cisplatin model of acute kidney injury, which can be linked to alterations in cell structure and transport function. In summary, we describe a powerful new tool to investigate how single cell behavior is integrated with whole organ structure and function and how it is altered in disease states relevant to humans.


Asunto(s)
Señalización del Calcio/fisiología , Calcio/metabolismo , Glomérulos Renales/metabolismo , Túbulos Renales Proximales/metabolismo , Lesión Renal Aguda/metabolismo , Animales , Humanos , Riñón/anatomía & histología , Riñón/metabolismo , Túbulos Renales Proximales/anatomía & histología , Ratones
6.
J Am Soc Nephrol ; 30(5): 737-750, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30902838

RESUMEN

BACKGROUND: A number of cAMP-elevating hormones stimulate phosphorylation (and hence activity) of the NaCl cotransporter (NCC) in the distal convoluted tubule (DCT). Evidence suggests that protein phosphatase 1 (PP1) and other protein phosphatases modulate NCC phosphorylation, but little is known about PP1's role and the mechanism regulating its function in the DCT. METHODS: We used ex vivo mouse kidney preparations to test whether a DCT-enriched inhibitor of PP1, protein phosphatase 1 inhibitor-1 (I1), mediates cAMP's effects on NCC, and conducted yeast two-hybrid and coimmunoprecipitation experiments in NCC-expressing MDCK cells to explore protein interactions. RESULTS: Treating isolated DCTs with forskolin and IBMX increased NCC phosphorylation via a protein kinase A (PKA)-dependent pathway. Ex vivo incubation of mouse kidney slices with isoproterenol, norepinephrine, and parathyroid hormone similarly increased NCC phosphorylation. The cAMP-induced stimulation of NCC phosphorylation strongly correlated with the phosphorylation of I1 at its PKA consensus phosphorylation site (a threonine residue in position 35). We also found an interaction between NCC and the I1-target PP1. Moreover, PP1 dephosphorylated NCC in vitro, and the PP1 inhibitor calyculin A increased NCC phosphorylation. Studies in kidney slices and isolated perfused kidneys of control and I1-KO mice demonstrated that I1 participates in the cAMP-induced stimulation of NCC. CONCLUSIONS: Our data suggest a complete signal transduction pathway by which cAMP increases NCC phosphorylation via a PKA-dependent phosphorylation of I1 and subsequent inhibition of PP1. This pathway might be relevant for the physiologic regulation of renal sodium handling by cAMP-elevating hormones, and may contribute to salt-sensitive hypertension in patients with endocrine disorders or sympathetic hyperactivity.


Asunto(s)
Transporte Biológico/efectos de los fármacos , Colforsina/farmacología , Túbulos Renales Distales/metabolismo , Proteína Fosfatasa 1/antagonistas & inhibidores , Proteínas/farmacología , Análisis de Varianza , Animales , Transporte Biológico/genética , Humanos , Immunoblotting , Técnicas In Vitro , Ratones , Ratones Noqueados , Fosforilación/efectos de los fármacos , Transducción de Señal/genética , Cloruro de Sodio/metabolismo , Miembro 3 de la Familia de Transportadores de Soluto 12/metabolismo
7.
J Am Soc Nephrol ; 29(7): 1849-1858, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29654216

RESUMEN

Background For many patients with kidney failure, the cause and underlying defect remain unknown. Here, we describe a novel mechanism of a genetic order characterized by renal Fanconi syndrome and kidney failure.Methods We clinically and genetically characterized members of five families with autosomal dominant renal Fanconi syndrome and kidney failure. We performed genome-wide linkage analysis, sequencing, and expression studies in kidney biopsy specimens and renal cells along with knockout mouse studies and evaluations of mitochondrial morphology and function. Structural studies examined the effects of recognized mutations.Results The renal disease in these patients resulted from monoallelic mutations in the gene encoding glycine amidinotransferase (GATM), a renal proximal tubular enzyme in the creatine biosynthetic pathway that is otherwise associated with a recessive disorder of creatine deficiency. In silico analysis showed that the particular GATM mutations, identified in 28 members of the five families, create an additional interaction interface within the GATM protein and likely cause the linear aggregation of GATM observed in patient biopsy specimens and cultured proximal tubule cells. GATM aggregates-containing mitochondria were elongated and associated with increased ROS production, activation of the NLRP3 inflammasome, enhanced expression of the profibrotic cytokine IL-18, and increased cell death.Conclusions In this novel genetic disorder, fully penetrant heterozygous missense mutations in GATM trigger intramitochondrial fibrillary deposition of GATM and lead to elongated and abnormal mitochondria. We speculate that this renal proximal tubular mitochondrial pathology initiates a response from the inflammasome, with subsequent development of kidney fibrosis.


Asunto(s)
Amidinotransferasas/genética , Síndrome de Fanconi/genética , Fallo Renal Crónico/genética , Mitocondrias/metabolismo , Mitocondrias/patología , Anciano , Amidinotransferasas/metabolismo , Animales , Simulación por Computador , Síndrome de Fanconi/complicaciones , Síndrome de Fanconi/metabolismo , Síndrome de Fanconi/patología , Femenino , Heterocigoto , Humanos , Lactante , Inflamasomas/metabolismo , Fallo Renal Crónico/etiología , Fallo Renal Crónico/metabolismo , Fallo Renal Crónico/patología , Masculino , Ratones , Ratones Noqueados , Conformación Molecular , Mutación , Mutación Missense , Linaje , Especies Reactivas de Oxígeno/metabolismo , Análisis de Secuencia de ADN , Adulto Joven
8.
EMBO Rep ; 14(12): 1143-8, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24157948

RESUMEN

Mechanical forces associated with fluid flow and/or circumferential stretch are sensed by renal epithelial cells and contribute to both adaptive or disease states. Non-selective stretch-activated ion channels (SACs), characterized by a lack of inactivation and a remarkably slow deactivation, are active at the basolateral side of renal proximal convoluted tubules. Knockdown of Piezo1 strongly reduces SAC activity in proximal convoluted tubule epithelial cells. Similarly, overexpression of Polycystin-2 (PC2) or, to a greater extent its pathogenic mutant PC2-740X, impairs native SACs. Moreover, PC2 inhibits exogenous Piezo1 SAC activity. PC2 coimmunoprecipitates with Piezo1 and deletion of its N-terminal domain prevents both this interaction and inhibition of SAC activity. These findings indicate that renal SACs depend on Piezo1, but are critically conditioned by PC2.


Asunto(s)
Células Epiteliales/metabolismo , Canales Iónicos/metabolismo , Mecanotransducción Celular , Canales Catiónicos TRPP/metabolismo , Potenciales de Acción , Animales , Sitios de Unión , Células COS , Células Cultivadas , Chlorocebus aethiops , Células Epiteliales/fisiología , Túbulos Renales/citología , Mutación , Unión Proteica , Canales Catiónicos TRPC/metabolismo , Canales Catiónicos TRPP/química , Canales Catiónicos TRPP/genética
9.
J Biol Chem ; 285(10): 7838-45, 2010 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-20056604

RESUMEN

The calcium-activated chloride channel anoctamin1 (ANO1; TMEM16A) is fundamental for the function of epithelial organs. Mice lacking ANO1 expression exhibit transport defects and a pathology similar to cystic fibrosis. They also show a general defect of epithelial electrolyte transport. Here we analyzed expression of all ten members (ANO1-ANO10) in a broad range of murine tissues and detected predominant expression of ANO1, 6, 7, 8, 9, 10 in epithelial tissues, while ANO2, 3, 4, 5 are common in neuronal and muscle tissues. When expressed in Fisher Rat Thyroid (FTR) cells, all ANO proteins localized to the plasma membrane but only ANO1, 2, 6, and 7 produced Ca(2+)-activated Cl(-) conductance, as analyzed by ATP-induced iodide quenching of YFP fluorescence. In contrast ANO9 and ANO10 suppressed baseline Cl(-) conductance and coexpression of ANO9 with ANO1 inhibited ANO1 activity. Patch clamping of ANO-expressing FRT cells indicated that apart from ANO1 also ANO6 and 10 produced chloride currents, albeit with very different Ca(2+) sensitivity and activation time. We conclude that each tissue expresses a set of anoctamins that form cell- and tissue-specific Ca(2+)-dependent Cl(-) channels.


Asunto(s)
Calcio/metabolismo , Canales de Cloruro/metabolismo , Cloruros/metabolismo , Epitelio/metabolismo , Isoformas de Proteínas/metabolismo , Animales , Transporte Biológico/fisiología , Línea Celular , Membrana Celular/metabolismo , Humanos , Activación del Canal Iónico/fisiología , Ionomicina/metabolismo , Ionóforos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Técnicas de Placa-Clamp , Isoformas de Proteínas/genética , Ratas , Distribución Tisular
10.
J Biol Chem ; 285(4): 2642-52, 2010 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-19940143

RESUMEN

Anti-inflammatory effects of glucocorticoids (GCs) are partly mediated by up-regulation of DUSP1 (dual specificity phosphatase 1), which dephosphorylates and inactivates mitogen-activated protein kinases. We identified putative GC-responsive regions containing GC receptor (GR) binding site consensus sequences that are well conserved between human and mouse DUSP1 loci in position, orientation, and sequence (at least 11 of 15 positions identical) and lie within regions of extended sequence conservation (minimum 65% identity over at least 100 bp). These were located approximately 29, 28, 24, 4.6, and 1.3 kb upstream of the DUSP1 transcription start site. The homology-based approach successfully identified four cis-acting regions that mediated transcriptional responses to dexamethasone. However, there was surprising interspecies divergence in site usage. This could not be explained by variations of the GR binding sites themselves. Instead, variations in flanking sequences appear to have driven the evolutionary divergence in mechanisms of regulation of mouse and human DUSP1 genes. There was a good correlation between the ability of cis-acting elements to respond to GC in transiently transfected reporter constructs and their ability to recruit GR in the context of intact chromatin. We propose that divergence of gene regulation has involved the loss or gain of binding sites for accessory transcription factors that assist in GR recruitment. Finally, a novel GC-responsive region of the human DUSP1 gene contains a highly unusual element, in which three closely spaced GR half-sites are required for potent transcriptional activation by GC.


Asunto(s)
Dexametasona/farmacología , Fosfatasa 1 de Especificidad Dual/genética , Evolución Molecular , Glucocorticoides/farmacología , Activación Transcripcional/fisiología , Animales , Secuencia de Bases , Células COS , Chlorocebus aethiops , Dimerización , Fosfatasa 1 de Especificidad Dual/metabolismo , Fibroblastos/citología , Células HeLa , Humanos , Ratones , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Regiones Promotoras Genéticas/fisiología , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Elementos de Respuesta/fisiología , Especificidad de la Especie , Activación Transcripcional/efectos de los fármacos
11.
Biol Chem ; 392(1-2): 125-34, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21194364

RESUMEN

Two families of proteins, the bestrophins (Best) and the recently cloned TMEM16 proteins (anoctamin, Ano), recapitulate properties of Ca(2+)-activated Cl(-) currents. Best1 is strongly expressed in the retinal pigment epithelium and could have a function as a Ca(2+)-activated Cl(-) channel as well as a regulator of Ca(2+) signaling. It is also present at much lower levels in other cell types including epithelial cells, where it regulates plasma membrane localized Cl(-) channels by controlling intracellular Ca(2+) levels. Best1 interacts with important Ca(2+)-signaling proteins such as STIM1 and can interact directly with other Ca(2+)-activated Cl(-) channels such as TMEM16A. Best1 is detected in the endoplasmic reticulum (ER) where it shapes the dynamic ER structure and regulates cell proliferation, which could be important for renal cystogenesis. Ca(2+)-activated Cl(-) channels of the anoctamin family (TMEM16A) show biophysical and pharmacological properties that are typical for endogenous Ca(2+)-dependent Cl(-) channels. TMEM16 proteins are abundantly expressed and many reports demonstrate their physiological importance in epithelial as well as non-epithelial cells. These channels are also activated by cell swelling and can therefore control cell volume, proliferation and apoptosis. To fully understand the function and regulation of Ca(2+)-activated Cl(-) currents, it is necessary to appreciate that Best1 and TMEM16A are embedded in a protein network and that they probably operate in functional microdomains.


Asunto(s)
Canales de Cloruro/metabolismo , Células Epiteliales/metabolismo , Proteínas del Ojo/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Anoctamina-1 , Bestrofinas , Células Epiteliales/química , Humanos
12.
J Biol Chem ; 284(42): 28698-703, 2009 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-19679661

RESUMEN

Molecular identification of the Ca(2+)-dependent chloride channel TMEM16A (ANO1) provided a fundamental step in understanding Ca(2+)-dependent Cl(-) secretion in epithelia. TMEM16A is an intrinsic constituent of Ca(2+)-dependent Cl(-) channels in cultured epithelia and may control salivary output, but its physiological role in native epithelial tissues remains largely obscure. Here, we demonstrate that Cl(-) secretion in native epithelia activated by Ca(2+)-dependent agonists is missing in mice lacking expression of TMEM16A. Ca(2+)-dependent Cl(-) transport was missing or largely reduced in isolated tracheal and colonic epithelia, as well as hepatocytes and acinar cells from pancreatic and submandibular glands of TMEM16A(-/-) animals. Measurement of particle transport on the surface of tracheas ex vivo indicated largely reduced mucociliary clearance in TMEM16A(-/-) mice. These results clearly demonstrate the broad physiological role of TMEM16A(-/-) for Ca(2+)-dependent Cl(-) secretion and provide the basis for novel treatments in cystic fibrosis, infectious diarrhea, and Sjöegren syndrome.


Asunto(s)
Canales de Cloruro/química , Cloruros/metabolismo , Animales , Anoctamina-1 , Calcio/química , Canales de Cloruro/metabolismo , Cloruros/química , Colon/patología , AMP Cíclico/metabolismo , Electrólitos , Células Epiteliales/citología , Hepatocitos/citología , Transporte Iónico , Ratones , Ratones Noqueados , Tráquea/citología
13.
J Biol Chem ; 283(38): 25765-9, 2008 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-18541529

RESUMEN

The powerful anti-inflammatory effects of glucocorticoids (GCs) have been known for more than sixty years, but their molecular mechanisms are still incompletely understood and hotly debated. The GC receptor (GR) was cloned in 1985 and shown to be a transcription factor. Initially, the anti-inflammatory actions of GCs were explained in terms of genes that were up-regulated by the receptor. However, none of these putative mediators seemed able to account for the spectrum of anti-inflammatory responses to GCs. The discovery of a negative regulatory function of GR then shifted the focus away from GC-induced genes as anti-inflammatory mediators. In recent years, attention has begun to move back toward the idea that the anti-inflammatory response to GCs is partially dependent on the positive regulation of gene expression by GR.


Asunto(s)
Fosfatasas de Especificidad Dual/fisiología , Regulación de la Expresión Génica , Glucocorticoides/metabolismo , Animales , Antiinflamatorios/farmacología , Fosfatasas de Especificidad Dual/genética , Humanos , Inflamación , Ligandos , Ratones , Modelos Biológicos , Receptores de Glucocorticoides/metabolismo , Transducción de Señal , Activación Transcripcional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA