Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Thorax ; 75(12): 1089-1094, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32917840

RESUMEN

OBJECTIVE: To determine the rates of asymptomatic viral carriage and seroprevalence of SARS-CoV-2 antibodies in healthcare workers. DESIGN: A cross-sectional study of asymptomatic healthcare workers undertaken on 24/25 April 2020. SETTING: University Hospitals Birmingham NHS Foundation Trust (UHBFT), UK. PARTICIPANTS: 545 asymptomatic healthcare workers were recruited while at work. Participants were invited to participate via the UHBFT social media. Exclusion criteria included current symptoms consistent with COVID-19. No potential participants were excluded. INTERVENTION: Participants volunteered a nasopharyngeal swab and a venous blood sample that were tested for SARS-CoV-2 RNA and anti-SARS-CoV-2 spike glycoprotein antibodies, respectively. Results were interpreted in the context of prior illnesses and the hospital departments in which participants worked. MAIN OUTCOME MEASURE: Proportion of participants demonstrating infection and positive SARS-CoV-2 serology. RESULTS: The point prevalence of SARS-CoV-2 viral carriage was 2.4% (n=13/545). The overall seroprevalence of SARS-CoV-2 antibodies was 24.4% (n=126/516). Participants who reported prior symptomatic illness had higher seroprevalence (37.5% vs 17.1%, χ2=21.1034, p<0.0001) and quantitatively greater antibody responses than those who had remained asymptomatic. Seroprevalence was greatest among those working in housekeeping (34.5%), acute medicine (33.3%) and general internal medicine (30.3%), with lower rates observed in participants working in intensive care (14.8%). BAME (Black, Asian and minority ethnic) ethnicity was associated with a significantly increased risk of seropositivity (OR: 1.92, 95% CI 1.14 to 3.23, p=0.01). Working on the intensive care unit was associated with a significantly lower risk of seropositivity compared with working in other areas of the hospital (OR: 0.28, 95% CI 0.09 to 0.78, p=0.02). CONCLUSIONS AND RELEVANCE: We identify differences in the occupational risk of exposure to SARS-CoV-2 between hospital departments and confirm asymptomatic seroconversion occurs in healthcare workers. Further investigation of these observations is required to inform future infection control and occupational health practices.


Asunto(s)
Anticuerpos Antivirales/sangre , Enfermedades Asintomáticas , COVID-19/diagnóstico , Personal de Salud/estadística & datos numéricos , Pandemias , SARS-CoV-2/inmunología , Adulto , COVID-19/epidemiología , COVID-19/virología , Estudios Transversales , Femenino , Humanos , Masculino , Persona de Mediana Edad , ARN Viral/análisis , SARS-CoV-2/genética , Estudios Seroepidemiológicos
2.
J Clin Virol ; 128: 104469, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32474371

RESUMEN

BACKGROUND: In January 2020 reports of unidentified severe respiratory illness were described in Wuhan, China. A rapid expansion in cases affecting most countries around the globe led to major changes in the way people live their daily lives. In the United Kingdom, the Department of Health and Social Care directed healthcare providers to establish additional resources to manage the anticipated surge in cases that could overwhelm the health services. A priority area was testing for SARS-CoV-2 RNA and its detection by qualitative RT-PCR. DESIGN: A laboratory workflow twinning research environment with clinical laboratory capabilities was implemented and validated in the University of Birmingham within 4 days of the project initiation. The diagnostic capability was centred on an IVD CE-marked RT-PCR kit and designed to provide surge capacity to the nearby Queen Elizabeth Hospital. The service was initially tasked with testing healthcare workers (HCW) using throat swabs, and subsequently the process investigated the utility of using saliva as an alternative sample type. RESULTS: Between the 8th April 2020 and the 30th April 2020, the laboratory tested a total of 1282 HCW for SARS-CoV-2 RNA in throat swabs. RNA was detected in 54 % of those who reported symptoms compatible with COVID-19, but in only 4% who were asymptomatic. CONCLUSION: This capability was established rapidly and utilised a cold-chain free methodology, applicable to a wide range of settings, and which can provide surge capacity and support to clinical laboratories facing increasing pressure during periods of national crisis.


Asunto(s)
Betacoronavirus/aislamiento & purificación , Técnicas de Laboratorio Clínico/métodos , Infecciones por Coronavirus/diagnóstico , Neumonía Viral/diagnóstico , ARN Viral/sangre , Betacoronavirus/genética , COVID-19 , Prueba de COVID-19 , Infecciones por Coronavirus/virología , Humanos , Pandemias , Neumonía Viral/virología , SARS-CoV-2 , Saliva/virología , Capacidad de Reacción , Reino Unido , Flujo de Trabajo
3.
Skelet Muscle ; 10(1): 5, 2020 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-32075690

RESUMEN

BACKGROUND: Hexose-6-Phosphate Dehydrogenase (H6PD) is a generator of NADPH in the Endoplasmic/Sarcoplasmic Reticulum (ER/SR). Interaction of H6PD with 11ß-hydroxysteroid dehydrogenase type 1 provides NADPH to support oxo-reduction of inactive to active glucocorticoids, but the wider understanding of H6PD in ER/SR NAD(P)(H) homeostasis is incomplete. Lack of H6PD results in a deteriorating skeletal myopathy, altered glucose homeostasis, ER stress and activation of the unfolded protein response. Here we further assess muscle responses to H6PD deficiency to delineate pathways that may underpin myopathy and link SR redox status to muscle wide metabolic adaptation. METHODS: We analysed skeletal muscle from H6PD knockout (H6PDKO), H6PD and NRK2 double knockout (DKO) and wild-type (WT) mice. H6PDKO mice were supplemented with the NAD+ precursor nicotinamide riboside. Skeletal muscle samples were subjected to biochemical analysis including NAD(H) measurement, LC-MS based metabolomics, Western blotting, and high resolution mitochondrial respirometry. Genetic and supplement models were assessed for degree of myopathy compared to H6PDKO. RESULTS: H6PDKO skeletal muscle showed adaptations in the routes regulating nicotinamide and NAD+ biosynthesis, with significant activation of the Nicotinamide Riboside Kinase 2 (NRK2) pathway. Associated with changes in NAD+ biosynthesis, H6PDKO muscle had impaired mitochondrial respiratory capacity with altered mitochondrial acylcarnitine and acetyl-CoA metabolism. Boosting NAD+ levels through the NRK2 pathway using the precursor nicotinamide riboside elevated NAD+/NADH but had no effect to mitigate ER stress and dysfunctional mitochondrial respiratory capacity or acetyl-CoA metabolism. Similarly, H6PDKO/NRK2 double KO mice did not display an exaggerated timing or severity of myopathy or overt change in mitochondrial metabolism despite depression of NAD+ availability. CONCLUSIONS: These findings suggest a complex metabolic response to changes in muscle SR NADP(H) redox status that result in impaired mitochondrial energy metabolism and activation of cellular NAD+ salvage pathways. It is possible that SR can sense and signal perturbation in NAD(P)(H) that cannot be rectified in the absence of H6PD. Whether NRK2 pathway activation is a direct response to changes in SR NAD(P)(H) availability or adaptation to deficits in metabolic energy availability remains to be resolved.


Asunto(s)
Músculo Esquelético/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Retículo Sarcoplasmático/metabolismo , Acetilcoenzima A/metabolismo , Animales , Deshidrogenasas de Carbohidratos/genética , Deshidrogenasas de Carbohidratos/metabolismo , Carnitina/análogos & derivados , Carnitina/metabolismo , Femenino , Masculino , Metaboloma , Ratones , Ratones Endogámicos C57BL , Mitocondrias Musculares/metabolismo , Niacinamida/análogos & derivados , Niacinamida/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Compuestos de Piridinio/metabolismo
4.
Cell Rep ; 21(12): 3498-3513, 2017 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-29262329

RESUMEN

Breast cancer progression, treatment resistance, and relapse are thought to originate from a small population of tumor cells, breast cancer stem cells (BCSCs). Identification of factors critical for BCSC function is therefore vital for the development of therapies. Here, we identify the arginine methyltransferase PRMT5 as a key in vitro and in vivo regulator of BCSC proliferation and self-renewal and establish FOXP1, a winged helix/forkhead transcription factor, as a critical effector of PRMT5-induced BCSC function. Mechanistically, PRMT5 recruitment to the FOXP1 promoter facilitates H3R2me2s, SET1 recruitment, H3K4me3, and gene expression. Our findings are clinically significant, as PRMT5 depletion within established tumor xenografts or treatment of patient-derived BCSCs with a pre-clinical PRMT5 inhibitor substantially reduces BCSC numbers. Together, our findings highlight the importance of PRMT5 in BCSC maintenance and suggest that small-molecule inhibitors of PRMT5 or downstream targets could be an effective strategy eliminating this cancer-causing population.


Asunto(s)
Neoplasias de la Mama/genética , Factores de Transcripción Forkhead/genética , Código de Histonas , Células Madre Neoplásicas/metabolismo , Proteína-Arginina N-Metiltransferasas/genética , Proteínas Represoras/genética , Animales , Neoplasias de la Mama/metabolismo , Proliferación Celular , Inhibidores Enzimáticos/farmacología , Epigénesis Genética , Femenino , Factores de Transcripción Forkhead/metabolismo , Humanos , Células MCF-7 , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/fisiología , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas Represoras/metabolismo
5.
Cell Biochem Funct ; 35(5): 269-277, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28749080

RESUMEN

Glucocorticoids are important for skeletal muscle energy metabolism, regulating glucose utilization, insulin sensitivity, and muscle mass. Nicotinamide adenine dinucleotide phosphate-dependent 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1)-mediated glucocorticoid activation in the sarcoplasmic reticulum (SR) is integral to mediating the detrimental effects of glucocorticoid excess in muscle. 11ß-Hydroxysteroid dehydrogenase type 1 activity requires glucose-6-phosphate transporter (G6PT)-mediated G6P transport into the SR for its metabolism by hexose-6-phosphate dehydrogenase (H6PDH) for NADPH generation. Here, we examine the G6PT/H6PDH/11ß-HSD1 triad in differentiating myotubes and explore the consequences of muscle-specific knockout of 11ß-HSD1 and H6PDH. 11ß-Hydroxysteroid dehydrogenase type 1 expression and activity increase with myotube differentiation and in response to glucocorticoids. Hexose-6-phosphate dehydrogenase shows some elevation in expression with differentiation and in response to glucocorticoid, while G6PT appears largely unresponsive to these particular conditions. When examining 11ß-HSD1 muscle-knockout mice, we were unable to detect significant decrements in activity, despite using a well-validated muscle-specific Cre transgene and confirming high-level recombination of the floxed HSD11B1 allele. We propose that the level of recombination at the HSD11B1 locus may be insufficient to negate basal 11ß-HSD1 activity for a protein with a long half-life. Hexose-6-phosphate dehydrogenase was undetectable in H6PDH muscle-knockout mice, which display the myopathic phenotype seen in global KO mice, validating the importance of SR NADPH generation. We envisage these data and models finding utility when investigating the muscle-specific functions of the 11ß-HSD1/G6PT/H6PDH triad.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/genética , Antiportadores/genética , Deshidrogenasas de Carbohidratos/genética , Proteínas de Transporte de Monosacáridos/genética , Músculo Esquelético/metabolismo , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Animales , Antiportadores/metabolismo , Deshidrogenasas de Carbohidratos/metabolismo , Metabolismo Energético/genética , Glucocorticoides/genética , Glucocorticoides/metabolismo , Glucosa/metabolismo , Resistencia a la Insulina/genética , Ratones , Ratones Noqueados , Proteínas de Transporte de Monosacáridos/metabolismo , Retículo Sarcoplasmático/genética , Retículo Sarcoplasmático/metabolismo
6.
J Endocrinol ; 220(3): 389-96, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24413279

RESUMEN

The activity of the enzyme 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1), which converts inactive cortisone (11-dehydrocorticosterone (11-DHC)) (in mice) into the active glucocorticoid (GC) cortisol (corticosterone in mice), can amplify tissue GC exposure. Elevated TNFα is a common feature in a range of inflammatory disorders and is detrimental to muscle function in diseases such as rheumatoid arthritis and chronic obstructive pulmonary disease. We have previously demonstrated that 11ß-HSD1 activity is increased in the mesenchymal stromal cells (MSCs) by TNFα treatment and suggested that this is an autoregulatory anti-inflammatory mechanism. This upregulation was mediated by the P2 promoter of the Hsd11b1 gene and was dependent on the NF-κB signalling pathway. In this study, we show that in contrast to MSCs, in differentiated C2C12 and primary murine myotubes, TNFα suppresses Hsd11b1 mRNA expression and activity through the utilization of the alternative P1 promoter. As with MSCs, in response to TNFα treatment, NF-κB p65 was translocated to the nucleus. However, ChIP analysis demonstrated that the direct binding was seen at position -218 to -245 bp of the Hsd11b1 gene's P1 promoter but not at the P2 promoter. These studies demonstrate the existence of differential regulation of 11ß-HSD1 expression in muscle cells through TNFα/p65 signalling and the P1 promoter, further enhancing our understanding of the role of 11ß-HSD1 in the context of inflammatory disease.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/genética , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Unión Proteica , Transducción de Señal , Factor de Transcripción ReIA/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Núcleo Celular/genética , Núcleo Celular/metabolismo , Regulación hacia Abajo , Regulación Enzimológica de la Expresión Génica , Ratones , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/enzimología , Músculo Esquelético/citología , Músculo Esquelético/enzimología , Músculo Esquelético/metabolismo , Regiones Promotoras Genéticas , Transporte de Proteínas , Factor de Transcripción ReIA/genética , Factor de Necrosis Tumoral alfa/genética
7.
Drug Metab Dispos ; 41(9): 1671-8, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23804523

RESUMEN

Bupropion is widely used for treatment of depression and as a smoking-cessation drug. Despite more than 20 years of therapeutic use, its metabolism is not fully understood. While CYP2B6 is known to form hydroxybupropion, the enzyme(s) generating erythro- and threohydrobupropion have long remained unclear. Previous experiments using microsomal preparations and the nonspecific inhibitor glycyrrhetinic acid suggested a role for 11ß-hydroxysteroid dehydrogenase 1 (11ß-HSD1) in the formation of both erythro- and threohydrobupropion. 11ß-HSD1 catalyzes the conversion of inactive glucocorticoids (cortisone, prednisone) to their active forms (cortisol, prednisolone). Moreover, it accepts several other substrates. Here, we used for the first time recombinant 11ß-HSD1 to assess its role in the carbonyl reduction of bupropion. Furthermore, we applied human, rat, and mouse liver microsomes and a selective inhibitor to characterize species-specific differences and to estimate the relative contribution of 11ß-HSD1 to bupropion metabolism. The results revealed 11ß-HSD1 as the major enzyme responsible for threohydrobupropion formation. The reaction was stereoselective and no erythrohydrobupropion was formed. Human liver microsomes showed 10 and 80 times higher activity than rat and mouse liver microsomes, respectively. The formation of erythrohydrobupropion was not altered in experiments with microsomes from 11ß-HSD1-deficient mice or upon incubation with 11ß-HSD1 inhibitor, indicating the existence of another carbonyl reductase that generates erythrohydrobupropion. Molecular docking supported the experimental findings and suggested that 11ß-HSD1 selectively converts R-bupropion to threohydrobupropion. Enzyme inhibition experiments suggested that exposure to bupropion is not likely to impair 11ß-HSD1-dependent glucocorticoid activation but that pharmacological administration of cortisone or prednisone may inhibit 11ß-HSD1-dependent bupropion metabolism.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Bupropión/análogos & derivados , Bupropión/metabolismo , Anciano , Animales , Bupropión/farmacocinética , Línea Celular , Cortisona/metabolismo , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microsomas Hepáticos/enzimología , Microsomas Hepáticos/metabolismo , Oxidación-Reducción , Prednisona/farmacocinética , Ratas , Ratas Sprague-Dawley
8.
Biochem Pharmacol ; 85(9): 1370-8, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23419873

RESUMEN

11ß-Hydroxysteroid dehydrogenase 1 (11ß-HSD1) catalyzes the conversion of inactive 11-oxo glucocorticoids (endogenous cortisone, 11-dehydrocorticosterone and synthetic prednisone) to their potent 11ß-hydroxyl forms (cortisol, corticosterone and prednisolone). Besides, 11ß-HSD1 accepts several other substrates. Using rodent liver microsomes and the unspecific inhibitor glycyrrhetinic acid, it has been proposed earlier that 11ß-HSD1 catalyzes the reversible conversion of the fungicide triadimefon to triadimenol. In the present study, recombinant human, rat and mouse enzymes together with a highly selective 11ß-HSD1 inhibitor were applied to assess the role of 11ß-HSD1 in the reduction of triadimefon and to uncover species-specific differences. To further demonstrate the role of 11ß-HSD1 in the carbonyl reduction of triadimefon, microsomes from liver-specific 11ß-HSD1-deficient mice were employed. Molecular docking was applied to investigate substrate binding. The results revealed important species differences and demonstrated the irreversible 11ß-HSD1-dependent reduction of triadimefon. Human liver microsomes showed 4 and 8 times higher activity than rat and mouse liver microsomes. The apparent Vmax/Km of recombinant human 11ß-HSD1 was 5 and 15 times higher than that of mouse and rat 11ß-HSD1, respectively, indicating isoform-specific differences and different expression levels for the three species. Experiments using inhibitors and microsomes from 11ß-HSD1-deficient mice indicated that 11ß-HSD1 is the major if not only enzyme responsible for triadimenol formation. The IC50 values of triadimefon and triadimenol for cortisone reduction suggested that exposure to these xenobiotica unlikely impairs the 11ß-HSD1-dependent glucocorticoid activation. However, elevated glucocorticoids during stress or upon pharmacological administration likely inhibit 11ß-HSD1-dependent metabolism of triadimefon in humans.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Fungicidas Industriales/farmacocinética , Triazinas/farmacocinética , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/antagonistas & inhibidores , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/química , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/metabolismo , Animales , Cortisona/metabolismo , Fungicidas Industriales/química , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microsomas Hepáticos/metabolismo , Modelos Moleculares , Oxidación-Reducción , Unión Proteica , Ratas , Ratas Sprague-Dawley , Especificidad de la Especie , Triazinas/química , Triazoles/química , Triazoles/metabolismo
9.
Endocrinology ; 153(7): 3236-48, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22555437

RESUMEN

Glucocorticoids (GC) are implicated in the development of metabolic syndrome, and patients with GC excess share many clinical features, such as central obesity and glucose intolerance. In patients with obesity or type 2 diabetes, systemic GC concentrations seem to be invariably normal. Tissue GC concentrations determined by the hypothalamic-pituitary-adrenal (HPA) axis and local cortisol (corticosterone in mice) regeneration from cortisone (11-dehydrocorticosterone in mice) by the 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1) enzyme, principally expressed in the liver. Transgenic mice have demonstrated the importance of 11ß-HSD1 in mediating aspects of the metabolic syndrome, as well as HPA axis control. In order to address the primacy of hepatic 11ß-HSD1 in regulating metabolism and the HPA axis, we have generated liver-specific 11ß-HSD1 knockout (LKO) mice, assessed biomarkers of GC metabolism, and examined responses to high-fat feeding. LKO mice were able to regenerate cortisol from cortisone to 40% of control and had no discernible difference in a urinary metabolite marker of 11ß-HSD1 activity. Although circulating corticosterone was unaltered, adrenal size was increased, indicative of chronic HPA stimulation. There was a mild improvement in glucose tolerance but with insulin sensitivity largely unaffected. Adiposity and body weight were unaffected as were aspects of hepatic lipid homeostasis, triglyceride accumulation, and serum lipids. Additionally, no changes in the expression of genes involved in glucose or lipid homeostasis were observed. Liver-specific deletion of 11ß-HSD1 reduces corticosterone regeneration and may be important for setting aspects of HPA axis tone, without impacting upon urinary steroid metabolite profile. These discordant data have significant implications for the use of these biomarkers of 11ß-HSD1 activity in clinical studies. The paucity of metabolic abnormalities in LKO points to important compensatory effects by HPA activation and to a crucial role of extrahepatic 11ß-HSD1 expression, highlighting the contribution of cross talk between GC target tissues in determining metabolic phenotype.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/genética , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Regulación Enzimológica de la Expresión Génica , Hígado/metabolismo , Alelos , Animales , Biomarcadores/metabolismo , Cortisona/análogos & derivados , Cortisona/farmacología , Glucocorticoides/metabolismo , Hidrocortisona/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microsomas Hepáticos/metabolismo , Fenotipo
10.
Mol Cell Endocrinol ; 336(1-2): 213-8, 2011 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-21146583

RESUMEN

Hexose-6-phosphate dehydrogenase (H6PDH) has emerged as an important factor in setting the redox status of the endoplasmic reticulum (ER) lumen. An important role of H6PDH is to generate a high NADPH/NADP(+) ratio which permits 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1) to act as an oxo-reductase, catalyzing the activation of glucocorticoids (GCs). In H6PDH knockout mice 11ß-HSD1 assumes dehydrogenase activity and inactivates GCs, rendering the target cell relatively GC insensitive. Consequently, H6PDHKO mice have a phenotype consistent with defects in the permissive and adaptive actions of GCs upon physiology. H6PDHKO mice have also offered an insight into muscle physiology as they also present with a severe vacuolating myopathy, abnormalities of glucose homeostasis and activation of the unfolded protein response due to ER stress, and a number of mechanisms driving this phenotype are thought to be involved. This article will review what we understand of the redox control of GC hormone metabolism regulated by H6PDH, and how H6PDHKO mice have allowed an in-depth understanding of its potentially novel, GC-independent roles in muscle physiology.


Asunto(s)
Deshidrogenasas de Carbohidratos/deficiencia , Animales , Deshidrogenasas de Carbohidratos/metabolismo , Retículo Endoplásmico/enzimología , Glucocorticoides/metabolismo , Humanos , Ratones , Ratones Noqueados , Enfermedades Musculares/enzimología , Enfermedades Musculares/patología , Enfermedades Musculares/fisiopatología , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...