Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Adv Neonatal Care ; 22(4): 291-299, 2022 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-34966056

RESUMEN

BACKGROUND: Extreme preterm infants face lengthy hospitalizations and are often subjected to painful stimuli. These stimuli may be related to routine caregiving that may negatively impact long-term developmental outcomes. Frequently obtained cuff blood pressure (BP) measurements are an example of a potentially noxious stimulus to preterm infants that may have a cumulating impact on development. PURPOSE: The primary aim was to explore the frequency of cuff BP measurements obtained in hemodynamically stable extreme preterm infants in the neonatal intensive care unit (NICU). Our secondary aim was to reduce the number of cuff BP measurements obtained in hemodynamically stable extreme preterm infants in the NICU. METHODS: Quality improvement methodologies per the Institute for Healthcare Improvement were used combined with a multidisciplinary approach. Participants were infants born less than 27 weeks of gestation and discharged home. The baseline period was 2015 through Q2-2018 and the intervention period was Q3-2018 through Q1-2020. The electronic medical record was used to collect data and Minitab Statistical Software was used for data analysis. FINDINGS/RESULTS: A baseline of 5.0% of eligible patients received the desired number of cuff BP measurements and increased to 63.2% after the intervention period. IMPLICATIONS FOR PRACTICE: Findings demonstrate that using quality improvement methodology can improve clinical care. Findings suggest the feasibility and safety of reducing the number of cuff BP measurements obtained on hemodynamically stable infants in the NICU. IMPLICATIONS FOR RESEARCH: Future endeavors should aim to reduce the quantity of painful stimuli in the NICU. Long-term developmental outcomes should be correlated in these patients.


Asunto(s)
Recien Nacido Prematuro , Unidades de Cuidado Intensivo Neonatal , Presión Sanguínea , Humanos , Lactante , Recién Nacido , Alta del Paciente , Mejoramiento de la Calidad
2.
J Christ Nurs ; 38(1): 32-37, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33284215

RESUMEN

Mercy is a valued nursing attribute. It often identifies faith-based educational institutions, clinics, and hospitals and has been a quality valued and taught by the Catholic Sisters of Mercy through nursing's history. This qualitative study explored the meaning of mercy as expressed by undergraduate nursing students attending a faith-based nursing school. This article presents the findings of a question posed to students: What does mercy mean to me? Nursing practice and education implications are also presented.


Asunto(s)
Catolicismo , Cristianismo , Empatía , Historia de la Enfermería , Atención de Enfermería/psicología , Filosofía en Enfermería/historia , Estudiantes de Enfermería/psicología , Adolescente , Adulto , Femenino , Historia del Siglo XVIII , Historia del Siglo XIX , Humanos , Masculino , Michigan , Medio Oeste de Estados Unidos , Investigación Cualitativa , Adulto Joven
3.
Toxicol Pathol ; 48(2): 362-378, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31640478

RESUMEN

Daprodustat (GSK1278863) is a hypoxia-inducible factor (HIF)-prolyl hydroxylase (PHD) inhibitor in development for treatment of anemia of chronic kidney disease. Daprodustat's biological activity simulates components of the natural response to hypoxia; inhibition of PHDs results in HIF stabilization and modulation of HIF-controlled gene products, including erythropoietin. The carcinogenic potential of daprodustat was evaluated in 2-year carcinogenicity studies in Sprague-Dawley rats and CD-1 mice, where once-daily doses were administered. The mouse study also included evaluation of daprodustat's 3 major circulating human metabolites. There were no neoplastic findings that were considered treatment related in either study. Exaggerated pharmacology resulted in significantly increased red cell mass and subsequent multiorgan congestion and secondary non-neoplastic effects in both species, similar to those observed in chronic toxicity studies. In rats, these included aortic thrombosis and an exacerbation of spontaneous rodent cardiomyopathy, which contributed to a statistically significant decrease in survival in high-dose males (group terminated in week 94). Survival was not impacted in mice at any dose. Systemic exposures (area under the plasma concentration-time curve) to daprodustat at the high doses in rats and mice exceed predicted maximal human clinical exposure by ≥143-fold. These results suggest that daprodustat and metabolites do not pose a carcinogenic risk at clinical doses.


Asunto(s)
Barbitúricos/toxicidad , Carcinogénesis/inducido químicamente , Pruebas de Carcinogenicidad , Evaluación Preclínica de Medicamentos , Glicina/análogos & derivados , Animales , Glicina/toxicidad , Prolina Dioxigenasas del Factor Inducible por Hipoxia/antagonistas & inhibidores , Ratones , Ratas , Ratas Sprague-Dawley
4.
Cancer Cell ; 36(1): 100-114.e25, 2019 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-31257072

RESUMEN

Type I protein arginine methyltransferases (PRMTs) catalyze asymmetric dimethylation of arginines on proteins. Type I PRMTs and their substrates have been implicated in human cancers, suggesting inhibition of type I PRMTs may offer a therapeutic approach for oncology. The current report describes GSK3368715 (EPZ019997), a potent, reversible type I PRMT inhibitor with anti-tumor effects in human cancer models. Inhibition of PRMT5, the predominant type II PRMT, produces synergistic cancer cell growth inhibition when combined with GSK3368715. Interestingly, deletion of the methylthioadenosine phosphorylase gene (MTAP) results in accumulation of the metabolite 2-methylthioadenosine, an endogenous inhibitor of PRMT5, and correlates with sensitivity to GSK3368715 in cell lines. These data provide rationale to explore MTAP status as a biomarker strategy for patient selection.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Purina-Nucleósido Fosforilasa/deficiencia , Empalme Alternativo , Antineoplásicos/química , Biomarcadores , Línea Celular Tumoral , Sinergismo Farmacológico , Inhibidores Enzimáticos/química , Humanos , Metilación , Modelos Moleculares , Conformación Molecular , Estructura Molecular , Unión Proteica , Proteína-Arginina N-Metiltransferasas/química , Especificidad por Sustrato
5.
J Pediatr Surg ; 54(11): 2428-2434, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30879741

RESUMEN

BACKGROUND: Skin-to-skin care (SSC) for infants improves physiologic stability, pain perception, brain development, parental bonding, and overall survival. Using quality improvement (QI) methodology, this project aimed to increase SSC for surgical infants in the neonatal intensive care unit (NICU). METHODS: A multidisciplinary working group composed of key NICU stakeholders instituted a needs assessment querying perceptions and concerns about SSC. Based on survey results, multiple system level interventions were implemented. Data for surgical infants receiving SSC during hospitalization were tracked over time using the electronic health record. RESULTS: Overall, 315 infants requiring a surgical consult were admitted to the NICU in the first 12 months of the project. After six months, SSC rates in this group increased from 51% to 60.5% (p < 0.01) and were sustained for 12 months. After one year, nursing staff reporting that they were somewhat to very comfortable providing SSC for surgical infants increased from 44% to 75% (p = 0.001) and the percent of nurses providing SSC for a surgical infant increased from 12% to 37% (p = 0.001). Inadvertent extubation did not significantly increase after implementation of the QI project. CONCLUSIONS: Using QI methodology and multidisciplinary engagement, SSC was integrated safely into the routine care of surgical infants in the NICU. LEVEL OF EVIDENCE: Level V.


Asunto(s)
Enfermedades del Recién Nacido/cirugía , Unidades de Cuidado Intensivo Neonatal/normas , Método Madre-Canguro , Estudios de Factibilidad , Encuestas de Atención de la Salud , Humanos , Recién Nacido , Mejoramiento de la Calidad
6.
PLoS One ; 12(1): e0169976, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28081568

RESUMEN

Systemic inflammation co-activates coagulation, which unchecked culminates in a lethal syndrome of multi-organ microvascular thrombosis known as disseminated intravascular coagulation (DIC). We studied an endotoxin-induced inflammatory state in rats to identify biomarkers of hemostatic imbalance favoring hypercoagulability. Intraperitoneal injection of LPS at 15 mg/kg body weight resulted in peripheral leukopenia and widespread neutrophilic sequestration characteristic of an acute systemic inflammatory response. Early indicators of hemostatic pathway activation developed within 4 hours, including increased circulating concentrations of procoagulant extracellular vesicles (EVs), EVs expressing endothelial cell and platelet membrane markers, and high concentration of soluble intercellular adhesion molecule-1 (sICAM-1), plasminogen activator inhibitor-1 (PAI-1), and D-dimers. Inflammation persisted throughout the 48-hour observation period; however, increases were found in a subset of serum microRNA (miRNA) that coincided with gradual resolution of hemostatic protein abnormalities and reduction in EV counts. Dose-adjusted LPS treatment in rats provides a time-course model to develop biomarker profiles reflecting procoagulant imbalance and rebalance under inflammatory conditions.


Asunto(s)
Lipopolisacáridos , Trombofilia/inducido químicamente , Trombofilia/fisiopatología , Enfermedad Aguda , Animales , Biomarcadores/sangre , Coagulación Sanguínea/efectos de los fármacos , Plaquetas/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Vesículas Extracelulares/metabolismo , Productos de Degradación de Fibrina-Fibrinógeno/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Leucopenia/inducido químicamente , Masculino , MicroARNs/sangre , Neutrófilos/metabolismo , Neutrófilos/patología , Inhibidor 1 de Activador Plasminogénico/metabolismo , Ratas , Ratas Wistar , Trombofilia/inmunología , Factores de Tiempo
7.
Toxicol Pathol ; 42(4): 635-57, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24777748

RESUMEN

Better biomarkers are needed to identify, characterize, and/or monitor drug-induced vascular injury (DIVI) in nonclinical species and patients. The Predictive Safety Testing Consortium (PSTC), a precompetitive collaboration of pharmaceutical companies and the U.S. Food and Drug Administration (FDA), formed the Vascular Injury Working Group (VIWG) to develop and qualify translatable biomarkers of DIVI. The VIWG focused its research on acute DIVI because early detection for clinical and nonclinical safety monitoring is desirable. The VIWG developed a strategy based on the premise that biomarkers of DIVI in rat would be translatable to humans due to the morphologic similarity of vascular injury between species regardless of mechanism. The histomorphologic lexicon for DIVI in rat defines degenerative and adaptive findings of the vascular endothelium and smooth muscles, and characterizes inflammatory components. We describe the mechanisms of these changes and their associations with candidate biomarkers for which advanced analytical method validation was completed. Further development is recommended for circulating microRNAs, endothelial microparticles, and imaging techniques. Recommendations for sample collection and processing, analytical methods, and confirmation of target localization using immunohistochemistry and in situ hybridization are described. The methods described are anticipated to aid in the identification and qualification of translational biomarkers for DIVI.


Asunto(s)
Biomarcadores/sangre , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Lesiones del Sistema Vascular/inducido químicamente , Lesiones del Sistema Vascular/patología , Animales , Evaluación Preclínica de Medicamentos/tendencias , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Humanos , Músculo Liso/efectos de los fármacos , Músculo Liso/patología , Estados Unidos , United States Food and Drug Administration
8.
Toxicol Pathol ; 41(1): 18-31, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22659243

RESUMEN

The measurement of plasma microRNAs (miRNAs) and messenger RNAs (mRNAs) is the most recent effort to identify novel biomarkers in preclinical safety. These genomic markers often display tissue-specific expression, may be released from the tissues into the plasma during toxic events, change early and with high magnitude in tissues and in the blood during specific organ toxicities, and can be measured using multiplex formats. Their validation as biomarkers has been challenged by the technical difficulties. In particular, the concentration of miRNAs in the plasma depends on contamination by miRNAs originating from blood cells and platelets, and the relative fraction of miRNAs in complexes with Argonaute 2, high-density lipoproteins, and in exosomes and microvesicles. In spite of these hurdles, considerable progress has recently been made in assessing the potential value of miRNAs in the clinic, especially in cancer patients and cardiovascular diseases. The future of miRNAs and mRNAs as biomarkers of disease and organ toxicity depends on our ability to characterize their kinetics and to establish robust collection and measurement methods. This review covers the basic biology of miRNAs and the published literature on the use of miRNAs and mRNAs as biomarkers of specific target organ toxicity.


Asunto(s)
MicroARNs/análisis , ARN Mensajero/análisis , Animales , Biomarcadores/análisis , Técnicas y Procedimientos Diagnósticos , Humanos
9.
Toxicol Appl Pharmacol ; 262(3): 310-20, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22627061

RESUMEN

Regulatory miRNAs play a role in vascular biology and are involved in biochemical and molecular pathways dysregulated during vascular injury. Collection and integration of functional miRNA data into these pathways can provide insight into pathogenesis at the site of injury; the same technologies applied to biofluids may provide diagnostic or surrogate biomarkers. miRNA was analyzed from mesentery and serum from rats given vasculotoxic compounds for 4 days. Fenoldopam, dopamine and midodrine each alter hemodynamics and are associated with histologic evidence of vascular injury, while yohimbine is vasoactive but does not cause histologic evidence of vascular injury in rat. There were 38 and 35 miRNAs altered in a statistically significant manner with a fold change of 2 or greater in mesenteries of fenoldopam- and dopamine-dosed rats, respectively, with 9 of these miRNAs shared. 10 miRNAs were altered in rats given midodrine; 6 were shared with either fenoldopam or dopamine. In situ hybridization demonstrated strong expression and co-localization of miR-134 in affected but not in adjacent unaffected vessels. Mesenteric miRNA expression may provide clarity or avenues of research into mechanisms involved in vascular injury once the functional role of specific miRNAs becomes better characterized. 102 miRNAs were altered in serum from rats with drug-induced vascular injury. 10 miRNAs were commonly altered in serum from dopamine and either fenoldopam or midodrine dosed rats; 18 of these 102 were also altered in mesenteries from rats with drug-induced vascular injury, suggesting their possible utility as peripheral biomarkers.


Asunto(s)
Vasos Sanguíneos/efectos de los fármacos , Mesenterio/metabolismo , Animales , Vasos Sanguíneos/metabolismo , Dopamina/farmacología , Fenoldopam/farmacología , Hemodinámica/efectos de los fármacos , Hibridación in Situ , Masculino , Mesenterio/efectos de los fármacos , MicroARNs/sangre , MicroARNs/genética , MicroARNs/metabolismo , Midodrina/farmacología , Ratas , Ratas Sprague-Dawley
10.
J Pharmacol Toxicol Methods ; 60(3): 263-74, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19577656

RESUMEN

INTRODUCTION: There are currently few widely accepted noninvasive detection methods for drug-induced vascular damage. Circulating endothelial progenitor cell (EPC) enumeration in humans has recently gained attention as a potential biomarker of vascular injury/endothelial damage/dysfunction. The rat is commonly used in preclinical drug development toxicity testing and lacks consensus noninvasive methodologies for immunophenotypic identification of EPCs. Identification of immunophenotypic markers of EPCs in the rat would enable transfer of technologies used in human for potential development of biomarkers for vascular injury the rat. Therefore, the aim of this work was to develop methods to consistently identify a discreet population of EPCs from rat peripheral blood. METHODS: EPCs were identified phenotypically from rat blood using cell culture, immunolabeling, fluorescence microscopy, and flow cytometry. EPCs isolated using immunolabeling coupled with magnetic separation and flow cytometric cell sorting were characterized genotypically using mRNA analysis. RESULTS: A modified colony forming unit (CFU)-Hill assay confirmed existence of immature EPCs in peripheral blood. Extended in vitro culture resulted in a morphology and immunophenotype consistent with mature endothelial cells as noted by positive staining for CD31, von Willebrand factor, rat endothelial cell antigen, and negative staining for smooth muscle cell alpha-actin. The majority of the cells identified as LDL+/CD11b/c(-) did not stain positively for either vWF or CD31. EPC populations isolated using magnetic separation and cell sorting were consistently positive for PECAM1, EDN1, FLK1, VWF, ITGAD, CCR1, IP30, and MMP2 mRNA expression. Cells identified as EPCs express cell-surface and gene expression markers consistent with endothelial cells and endothelial progenitor cell populations. DISCUSSION: Vascular trauma induces transient mobilization of EPCs in humans and their enumeration and characterization have been proposed as a surrogate biomarker for assessment of vascular injury. Potential exists for using rat circulating EPCs as a surrogate sampling population for biomarker development in drug-related injury in preclinical toxicity studies. A prerequisite to biomarker development is the ability to consistently identify a discreet population of EPCs from peripheral rat blood. This work describes novel methods for isolation and validation of phenotypically and genotypically consistent populations of rat EPCs from peripheral blood. These methods are well suited for potential future use in validation of enumeration and/or biomarker development methods in the rat.


Asunto(s)
Células Endoteliales/citología , Células Endoteliales/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Células Madre/citología , Células Madre/metabolismo , Animales , Biomarcadores/sangre , Células Cultivadas , Células Endoteliales/fisiología , Endotelio Vascular/fisiología , Humanos , Inmunofenotipificación , Masculino , Ratas , Ratas Sprague-Dawley , Células Madre/fisiología
11.
Toxicol Pathol ; 35(7): 958-71, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18098042

RESUMEN

Novel vascular lesions were observed in mice given an alpha vbeta 3, alpha vbeta 5 receptor antagonist (SB-273005) for up to 3 months. Vascular smooth muscle cell (VSMC) necrosis was observed in aorta and renal hilar arteries approximately 6 hours after dosing followed by loss of VSMC, adaptive medial thickening by VSMC hypertrophy and deposition of PAS-positive matrix and collagen. Renal hilar and arcuate arteries developed delayed and transient fibrinoid necrosis and inflammation. Vascular regeneration was not evident following drug-withdrawal after 3 days of dosing. Vascular lesions were associated with necrosis, regeneration and fibrosis of heart, kidney and spleen consistent with initial ischemic injury followed by tissue repair. VSMC toxicity was likely not related to integrin antagonism because lesions were not observed with related compounds and no vascular changes were observed in other preclinical species. In vitro studies failed to demonstrate a direct toxic effect of SB-273005 on VSMC or unique species sensitivity of murine VSMC. In conclusion, SB-273005 caused VSMC necrosis in aorta and renal arteries of mice. Lesions did not progress or recover, but there was medial hypertrophic adaptation even with continued dosing. This is considered direct species-specific VSMC toxicity of unknown mechanism and unrelated to vitronectin receptor antagonism.


Asunto(s)
Aorta/efectos de los fármacos , Integrina alfaVbeta3/antagonistas & inhibidores , Músculo Liso Vascular/efectos de los fármacos , Piridinas/toxicidad , Arteria Renal/efectos de los fármacos , Animales , Aorta/patología , Apoptosis/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Femenino , Corazón/efectos de los fármacos , Inmunohistoquímica , Riñón/efectos de los fármacos , Riñón/patología , Masculino , Ratones , Ratones Endogámicos ICR , Microscopía Electrónica , Músculo Liso Vascular/patología , Arteria Renal/patología , Bazo/efectos de los fármacos , Bazo/patología
12.
Toxicol Pathol ; 35(2): 284-95, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17366323

RESUMEN

TGF-beta, and its type 1 (ALK5) receptor, are critical to the pathogenesis of fibrosis. In toxicologic studies of 4 or more days in 10-week-old Sprague-Dawley rats, using an ALK5 inhibitor (GW788388), expansion of hypertrophic and proliferation zones of femoral physes were noted. Subphyseal hyperostosis, chondrocyte hypertrophy/hyperplasia, and increased matrix were present. Physeal zones were laser microdissected from ALK5 inhibitor-treated and control rats sacrificed after 3 days of treatment. Transcripts for TGF-beta1, TGF-beta2, ALK5, IHH, VEGF, BMP-7, IGF-1, bFGF, and PTHrP were amplified by real-time PCR. IGF and IHH increased in all physis zones with treatment, but were most prominent in prehypertrophic zones. TGF-beta2, bFGF and BMP7 expression increased in proliferative, pre-and hypertrophic zones. PTHrP expression was elevated in proliferative zones but decreased in hypertrophic zones. VEGF expression was increased after treatment in pre- and hypertrophic zones. ALK5 expression was elevated in prehypertrophic zones. Zymography demonstrated gelatinolytic activity was reduced after treatment. Apoptotic markers (TUNEL and caspase-3) were decreased in hypertrophic zones. Proliferation assessed by Topoisomerase II and Ki67 was increased in multiple zones. Movat stains demonstrated that proteoglycan deposition was altered. Physeal changes occurred at doses well above those resulting in fibrosis. Interactions of factors is important in producing the physeal dysplasia phenotype.


Asunto(s)
Receptores de Activinas Tipo I/metabolismo , Enfermedades del Desarrollo Óseo/inducido químicamente , Placa de Crecimiento/patología , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal/fisiología , Receptores de Activinas Tipo I/antagonistas & inhibidores , Receptores de Activinas Tipo I/genética , Animales , Benzamidas/efectos adversos , Enfermedades del Desarrollo Óseo/patología , Proliferación Celular , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica , Placa de Crecimiento/efectos de los fármacos , Proteínas Serina-Treonina Quinasas , Pirazoles/efectos adversos , Ratas , Ratas Sprague-Dawley , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Receptores de Factores de Crecimiento Transformadores beta/genética , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/fisiología , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/fisiología
13.
J Histochem Cytochem ; 54(11): 1229-37, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16864893

RESUMEN

Microarrays have been used to simultaneously monitor the expression of thousands of genes from biological samples, an approach that can potentially uncover previously unrecognized functions of genes. Microarray analyses can rarely be conducted retrospectively because of the requirement for RNA to be obtained from fresh or unfixed frozen tissues. Archived pathology specimens would need to be used for retrospective analyses, and these are typically preserved as formalin-fixed, paraffin-embedded (FFPE) tissue. Formalin-fixed tissues have been shown to yield compromised RNA compared with that obtained from frozen tissue. To begin to assess the performance of RNA extracted from FFPE samples on a microarray format, we compared RNA from a model system of pelleted lipopolysaccharide-stimulated human bone marrow stromal cells that were snap frozen with RNA from FFPE cells. RNA integrity and Affymetrix quality control parameters were assessed, and differentially regulated genes were analyzed with Ingenuity Pathway Analysis software. Results demonstrate that both snap-frozen and FFPE samples yielded intact RNA suitable for amplification prior to Affymetrix GeneChip analysis. Although some transcriptional information was lost with RNA extracted from the FFPE samples, Ingenuity Pathway Analysis revealed that the major pathways identified as affected by drug treatment were similar. Results show that FFPE samples are amenable to Affymetrix GeneChip analysis, expanding the possibility for expression profiling on archived tissue blocks in pathology laboratories.


Asunto(s)
Criopreservación , Fijadores , Formaldehído , Perfilación de la Expresión Génica , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Adhesión en Parafina , ARN/análisis , Células de la Médula Ósea/química , Células Cultivadas , Humanos , Lipopolisacáridos/farmacología , Control de Calidad , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células del Estroma/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...