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.
Endocr Relat Cancer ; 24(10): 519-529, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28830934

RESUMEN

Hyperinsulinemia is associated with a decrease in breast cancer recurrence-free survival and overall survival. Inhibition of insulin receptor signaling is associated with glycemic dysregulation. SET is a direct modulator of PP2A, which negatively regulates the PI3K/AKT/mTOR pathway. OP449, a SET inhibitor, decreases AKT/mTOR activation. The effects of OP449 treatment on breast cancer growth in the setting of pre-diabetes, and its metabolic implications are currently unknown. We found that the volumes and weights of human MDA-MB-231 breast cancer xenografts were greater in hyperinsulinemic mice compared with controls (P < 0.05), and IR phosphorylation was 4.5-fold higher in these mice (P < 0.05). Human and murine breast cancer tumors treated with OP449 were 47% and 39% smaller than controls (P < 0.05, for both, respectively). AKT and S6RP phosphorylation were 82% and 34% lower in OP449-treated tumors compared with controls (P < 0.05, P = 0.06, respectively). AKT and S6RP phosphorylation in response to insulin was 30% and 12% lower in cells, pre-treated with OP449, compared with control cells (P < 0.01, P < 0.05, respectively). However, even with decreased AKT/mTOR activation, body weights and composition, blood glucose and plasma insulin, glucose tolerance, serum triglyceride and cholesterol levels were similar between OP449-treated mice and controls. Xenografts and liver tissue from OP449-treated mice showed a 64% and 70% reduction in STAT5 activation, compared with controls (P < 0.01 and P = 0.06, respectively). Our data support an anti-neoplastic effect of OP449 on human breast cancer cells in vitro and in xenografts in the setting of hyperinsulinemia. OP449 led to the inhibition of AKT/mTOR signaling, albeit, not leading to metabolic derangements.


Asunto(s)
Neoplasias de la Mama/etiología , Complicaciones de la Diabetes , Hiperinsulinismo/complicaciones , Obesidad/complicaciones , Péptidos/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/mortalidad , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Análisis de Supervivencia
3.
Biol Reprod ; 95(5): 93, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27535958

RESUMEN

Endometriosis is an estrogen-dependent, progesterone-resistant disorder largely derived from retrograde transplantation of menstrual tissue/cells into the pelvis, eliciting an inflammatory response, pelvic pain, and infertility. Eutopic endometrium (within the uterus), giving rise to pelvic disease, displays cycle-dependent transcriptomic, proteomic, and signaling abnormalities, and although its DNA methylation profiles dynamically change across the cycle in healthy women, studies in endometriosis are limited. Herein, we investigated the DNA methylome and associated gene expression in three phases of the cycle in eutopic endometrium of women with severe endometriosis versus controls, matched for ethnicity, medications, smoking, and no recent contraceptive steroid use. Genome-wide DNA methylation and gene expression were coassessed in each sample. Cycle phase was determined by histology, serum hormone levels, and unsupervised principal component and hierarchical cluster analyses of microarray data. Altered endometrial DNA methylation in endometriosis was most prominent in the midsecretory phase (peak progesterone), with disruption of the normal pattern of cycle-dependent DNA methylation changes, including a bias toward methylation of CpG islands, suggesting wide-range abnormalities of the chromatin remodeling machinery in endometriosis. DNA methylation changes were associated with altered gene expression relevant to endometrial function/dysfunction, including cell proliferation, inflammation/immune response, angiogenesis, and steroid hormone response. The data provide insight into epigenetic reprogramming and steroid hormone actions in endometrium contributing to the pathogenesis and pathophysiology of endometriosis.


Asunto(s)
Metilación de ADN , Endometriosis/metabolismo , Endometrio/metabolismo , Expresión Génica , Ciclo Menstrual/metabolismo , Adulto , Islas de CpG , Endometriosis/genética , Epigénesis Genética , Femenino , Regulación de la Expresión Génica , Humanos , Ciclo Menstrual/genética , Proteómica
4.
Endocr Relat Cancer ; 23(9): 747-58, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27435064

RESUMEN

Type 2 diabetes (T2D) is associated with increased cancer risk and cancer-related mortality. Data herein show that we generated an immunodeficient hyperinsulinemic mouse by crossing the Rag1(-/-) mice, which have no mature B or T lymphocytes, with the MKR mouse model of T2D to generate the Rag1(-/-) (Rag/WT) and Rag1(-/-)/MKR(+/+) (Rag/MKR) mice. The female Rag/MKR mice are insulin resistant and have significantly higher nonfasting plasma insulin levels compared with the Rag/WT controls. Therefore, we used these Rag/MKR mice to investigate the role of endogenous hyperinsulinemia on human cancer progression. In this study, we show that hyperinsulinemia in the Rag/MKR mice increases the expression of mesenchymal transcription factors, TWIST1 and ZEB1, and increases the expression of the angiogenesis marker, vascular endothelial growth factor A (VEGFA). We also show that silencing the insulin receptor (IR) in the human LCC6 cancer cells leads to decreased tumor growth and metastases, suppression of mesenchymal markers vimentin, SLUG, TWIST1 and ZEB1, suppression of angiogenesis markers, VEGFA and VEGFD, and re-expression of the epithelial marker, E-cadherin. The data in this paper demonstrate that IR knockdown in primary tumors partially reverses the growth-promoting effects of hyperinsulinemia as well as highlighting the importance of the insulin receptor signaling pathway in cancer progression, and more specifically in epithelial-mesenchymal transition.


Asunto(s)
Transición Epitelial-Mesenquimal , Hiperinsulinismo/genética , Neoplasias Mamarias Experimentales/genética , Receptor de Insulina/genética , Animales , Línea Celular Tumoral , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patología , Modelos Animales de Enfermedad , Femenino , Silenciador del Gen , Humanos , Hiperinsulinismo/metabolismo , Hiperinsulinismo/patología , Masculino , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones Transgénicos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Receptor de Insulina/metabolismo , Transducción de Señal , Factores de Transcripción de la Familia Snail/genética , Factores de Transcripción de la Familia Snail/metabolismo , Proteína 1 Relacionada con Twist/genética , Proteína 1 Relacionada con Twist/metabolismo , Factor A de Crecimiento Endotelial Vascular , Vimentina/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo
5.
Diabetologia ; 59(9): 2018-25, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27241182

RESUMEN

AIMS/HYPOTHESIS: Previous epidemiological studies have reported a potential link between insulin analogues and breast cancer; however, a prospective randomised controlled trial showed neutral effects of insulin glargine on cancer risk. Insulin glargine is metabolised in vivo to an M1 metabolite. A question remains whether a subset of individuals with slower rates of glargine metabolism or who are on high doses could, theoretically, have an increased risk of cancer progression if a tumour is already present. In this study, we aimed to determine whether a non-metabolisable form of insulin glargine induced murine breast cancer growth. METHODS: A mouse model of type 2 diabetes (MKR) was used for these studies. MKR mice were injected with two murine mammary cancer cell lines: Mvt-1 cells (derived from MMTV-c-Myc/Vegf tumours) and Met1 cells (derived from MMTV-polyoma virus middle T antigen tumours). Mice were treated with 25 U/kg per day of the long-acting insulin analogues, insulin glargine, insulin detemir, insulin degludec or non-metabolisable glargine, or vehicle. RESULTS: No difference in tumour growth was seen in terms of tumour size after insulin glargine, detemir, degludec or vehicle injections. Non-metabolisable glargine did not increase tumour growth compared with insulin glargine or vehicle. Insulin glargine and non-metabolisable glargine led to insulin receptor phosphorylation in vivo rather than IGF-1 receptor phosphorylation. CONCLUSIONS/INTERPRETATION: These results demonstrate that in a mouse model of type 2 diabetes, at high concentrations, basal insulin analogues and a non-metabolisable glargine analogue do not promote the progression of breast tumours.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Insulina Glargina/efectos adversos , Neoplasias Mamarias Animales/inducido químicamente , Neoplasias Mamarias Animales/patología , Animales , Línea Celular Tumoral , Diabetes Mellitus Tipo 2/sangre , Modelos Animales de Enfermedad , Femenino , Humanos , Hipoglucemiantes/efectos adversos , Hipoglucemiantes/química , Hipoglucemiantes/uso terapéutico , Insulina/química , Insulina/uso terapéutico , Insulina Glargina/química , Insulina Glargina/uso terapéutico , Ratones , Fosforilación/efectos de los fármacos , Receptor de Insulina/metabolismo , Receptores de Somatomedina/metabolismo
6.
J Diabetes Complications ; 30(6): 999-1007, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27197688

RESUMEN

AIMS: Type 2 diabetes (T2D) is a complex metabolic disease leading to complications in multiple organs. Diabetic myopathy and liver disease are common complications of T2D, but are incompletely understood. To gain insight into the pathogenesis of these conditions we performed metabolomic analysis of skeletal muscle and liver in a mouse model of T2D. METHODS: Tissue metabolomics were performed by GC/MS and LC/MS of the skeletal muscle and liver in the MKR mouse model of T2D, compared with control mice. MKR mice were treated with the ß-3 adrenergic receptor agonist, CL-316,243 to determine metabolite changes after correcting hyperglycemia. RESULTS: Blood glucose was higher in MKR vs WT mice, and normalized with CL-316,243 treatment. Compared with WT mice, MKR mice had 2.5 fold higher concentrations of sorbitol and 1.7 fold lower concentrations of reduced glutathione in skeletal muscle. In liver, MKR mice had 2 fold higher concentrations of the pentitol ribitol. CL-316,243 treatment normalized sorbitol and ribitol concentrations in MKR skeletal muscle and liver, respectively to the levels of the WT mice. CONCLUSIONS: These results demonstrate tissue-specific accumulation of polyols in a mouse model of T2D and provide novel insights into the pathogenesis of myopathy and liver disease in T2D.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Hígado/metabolismo , Metaboloma , Músculo Esquelético/metabolismo , Polímeros/metabolismo , Animales , Diabetes Mellitus Experimental , Dioxoles/farmacología , Masculino , Ratones
7.
Mol Endocrinol ; 28(7): 1118-35, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24877562

RESUMEN

Human endometrium undergoes major gene expression changes, resulting in altered cellular functions in response to cyclic variations in circulating estradiol and progesterone, largely mediated by transcription factors and nuclear receptors. In addition to classic modulators, epigenetic mechanisms regulate gene expression during development in response to environmental factors and in some diseases and have roles in steroid hormone action. Herein, we tested the hypothesis that DNA methylation plays a role in gene expression regulation in human endometrium in different hormonal milieux. High throughput, genome-wide DNA methylation profiling of endometrial samples in proliferative, early secretory, and midsecretory phases revealed dynamic DNA methylation patterns with segregation of proliferative from secretory phase samples by unsupervised cluster analysis of differentially methylated genes. Changes involved different frequencies of gain and loss of methylation within or outside CpG islands. Comparison of changes in transcriptomes and corresponding DNA methylomes from the same samples revealed association of DNA methylation and gene expression in a number of loci, some important in endometrial biology. Human endometrial stromal fibroblasts treated in vitro with estradiol and progesterone exhibited DNA methylation changes in several genes observed in proliferative and secretory phase tissues, respectively. Taken together, the data support the observation that epigenetic mechanisms are involved in gene expression regulation in human endometrium in different hormonal milieux, adding endometrium to a small number of normal adult tissues exhibiting dynamic DNA methylation. The data also raise the possibility that the interplay between steroid hormone and methylome dynamics regulates normal endometrial functions and, if abnormal, may result in endometrial dysfunction and associated disorders.


Asunto(s)
Metilación de ADN , ADN/metabolismo , Endometrio/metabolismo , Epigénesis Genética/genética , Regulación de la Expresión Génica/genética , Adulto , Proliferación Celular , Análisis por Conglomerados , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Islas de CpG/genética , Receptor con Dominio Discoidina 1 , Endometrio/citología , Estradiol/sangre , Estradiol/farmacología , Femenino , Fibroblastos/citología , Perfilación de la Expresión Génica , Humanos , Metaloproteinasas de la Matriz Secretadas/genética , Proteínas de la Membrana/genética , Ciclo Menstrual/genética , Persona de Mediana Edad , Mucina-1/genética , Progesterona/sangre , Progesterona/farmacología , Proteínas Tirosina Quinasas Receptoras/genética , Células del Estroma/citología , Transcriptoma/genética
8.
Endocrinol Metab Clin North Am ; 43(1): 167-85, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24582097

RESUMEN

Diabetes is a worldwide health problem that has been increasingly associated with various types of cancers. Epidemiologic studies have shown an increased risk of cancer as well as a higher mortality rate in patients with type 2 diabetes (T2D). The biologic mechanisms driving the link between T2D and cancer are not well understood. In this review, various proposed mechanisms are addressed to explain the relationship between T2D and cancer. Understanding the precise mechanisms that link T2D, obesity, and the metabolic syndrome with cancer will aid in developing treatments that will reduce mortality in individuals with T2D and cancer.


Asunto(s)
Diabetes Mellitus Tipo 2/complicaciones , Neoplasias/etiología , Diabetes Mellitus Tipo 2/inmunología , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Neoplasias/inmunología , Neoplasias/metabolismo
9.
Diabetes ; 62(10): 3553-60, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23835331

RESUMEN

Endogenous hyperinsulinemia and insulin receptor (IR)/IGF-I receptor (IGF-IR) phosphorylation in tumors are associated with a worse prognosis in women with breast cancer. In vitro, insulin stimulation of the IR increases proliferation of breast cancer cells. However, in vivo studies demonstrating that IR activation increases tumor growth, independently of IGF-IR activation, are lacking. We hypothesized that endogenous hyperinsulinemia increases mammary tumor growth by directly activating the IR rather than the IGF-IR or hybrid receptors. We aimed to determine whether stimulating the IR with the insulin analog AspB10 could increase tumor growth independently of IGF-IR signaling. We induced orthotopic mammary tumors in control FVB/n and hyperinsulinemic MKR mice, and treated them with the insulin analog AspB10, recombinant human IGF-I, or vehicle. Tumors from mice with endogenous hyperinsulinemia were larger and had greater IR phosphorylation, but not IGF-IR phosphorylation, than those from control mice. Chronic AspB10 administration also increased tumor growth and IR (but not IGF-IR) phosphorylation in tumors. IGF-I led to activation of both the IGF-IR and IR and probably hybrid receptors. Our results demonstrate that IR phosphorylation increases tumor growth, independently of IGF-IR/hybrid receptor phosphorylation, and warrant consideration when developing therapeutics targeting the IGF-IR, but not the IR.


Asunto(s)
Neoplasias de la Mama/metabolismo , Diabetes Mellitus Experimental/tratamiento farmacológico , Hiperinsulinismo/complicaciones , Hipoglucemiantes/efectos adversos , Factor I del Crecimiento Similar a la Insulina/efectos de los fármacos , Insulina/análogos & derivados , Síndrome Metabólico/complicaciones , Terapia Molecular Dirigida , Obesidad/complicaciones , Receptor IGF Tipo 1/efectos de los fármacos , Animales , Neoplasias de la Mama/etiología , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Humanos , Hipoglucemiantes/administración & dosificación , Insulina/administración & dosificación , Insulina/efectos adversos , Neoplasias Mamarias Experimentales , Ratones , Ratones Transgénicos , Terapia Molecular Dirigida/tendencias , Fosforilación , Pronóstico , Transducción de Señal , Células Tumorales Cultivadas
10.
J Clin Endocrinol Metab ; 97(3): E376-92, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22259059

RESUMEN

CONTEXT: Endometriosis is characterized by progesterone resistance and associated with infertility. Krüppel-like factor 9 (KLF9) is a progesterone receptor (PGR)-interacting protein, and mice null for Klf9 are subfertile. Whether loss of KLF9 expression contributes to progesterone resistance of eutopic endometrium of women with endometriosis is unknown. OBJECTIVE: The aims were to investigate 1) KLF9 expression in eutopic endometrium of women with and without endometriosis, 2) effects of attenuated KLF9 expression on WNT-signaling component expression and on WNT inhibitor Dickkopf-1 promoter activity in human endometrial stromal cells (HESC), and 3) PGR and KLF9 coregulation of the stromal transcriptome network. METHODS: Transcript levels of KLF9, PGR, and WNT signaling components were measured in eutopic endometrium of women with and without endometriosis. Transcript and protein levels of WNT signaling components in HESC transfected with KLF9 and/or PGR small interfering RNA were analyzed by quantitative RT-PCR and Western blot. KLF9 and PGR coregulation of Dickkopf-1 promoter activity was evaluated using human Dickkopf-1-luciferase promoter/reporter constructs and by chromatin immunoprecipitation. KLF9 and PGR signaling networks were analyzed by gene expression array profiling. RESULTS: Eutopic endometrium from women with endometriosis had reduced expression of KLF9 mRNA together with those of PGR-B, WNT4, WNT2, and DKK1. KLF9 and PGR were recruited to the DKK1 promoter and modified each other's transactivity. In HESC, KLF9 and PGR coregulated components of the WNT, cytokine, and IGF gene networks that are implicated in endometriosis and infertility. CONCLUSION: Loss of KLF9 coregulation of endometrial stromal PGR-responsive gene networks may underlie progesterone resistance in endometriosis.


Asunto(s)
Endometriosis/metabolismo , Endometrio/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Receptores de Progesterona/metabolismo , Células del Estroma/metabolismo , Adulto , Endometriosis/etiología , Endometriosis/genética , Endometriosis/patología , Endometrio/patología , Femenino , Expresión Génica , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Regiones Promotoras Genéticas , Receptores de Progesterona/genética , Transducción de Señal/fisiología , Células del Estroma/patología , Vía de Señalización Wnt/fisiología
11.
J Assist Reprod Genet ; 29(2): 105-15, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22222853

RESUMEN

PURPOSE: To determine the vaginal microbiome in women undergoing IVF-ET and investigate correlations with clinical outcomes. METHODS: Thirty patients had blood drawn for estradiol (E(2)) and progesterone (P(4)) at four time points during the IVF-ET cycle and at 4-6 weeks of gestation, if pregnant. Vaginal swabs were obtained in different hormonal milieu, and the vaginal microbiome determined by deep sequencing of the 16S ribosomal RNA gene. RESULTS: The vaginal microbiome underwent a transition during therapy in some but not all patients. Novel bacteria were found in 33% of women tested during the treatment cycle, but not at 6-8 weeks of gestation. Diversity of species varied across different hormonal milieu, and on the day of embryo transfer correlated with outcome (live birth/no live birth). The species diversity index distinguished women who had a live birth from those who did not. CONCLUSIONS: This metagenomics approach has enabled discovery of novel, previously unidentified bacterial species in the human vagina in different hormonal milieu and supports a shift in the vaginal microbiome during IVF-ET therapy using standard protocols. Furthermore, the data suggest that the vaginal microbiome on the day of embryo transfer affects pregnancy outcome.


Asunto(s)
Bacterias/clasificación , Fertilización In Vitro , Metagenoma , ARN Ribosómico 16S/genética , Vagina/microbiología , Adulto , Transferencia de Embrión , Estradiol/sangre , Femenino , Humanos , Persona de Mediana Edad , Embarazo , Resultado del Embarazo , Progesterona/sangre
12.
Biol Reprod ; 86(2): 58, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22075475

RESUMEN

Human endometrium regenerates on a cyclic basis from candidate stem/progenitors whose genetic programs are yet to be determined. A subpopulation of endometrial stromal cells, displaying key properties of mesenchymal stem cells (MSCs), has been characterized. The endometrial MSC (eMSC) is likely the precursor of the endometrial stromal fibroblast. The goal of this study was to determine the transcriptome and signaling pathways in the eMSC to understand its functional phenotype. Endometrial stromal cells from oocyte donors (n = 20) and patients undergoing benign gynecologic surgery (n = 7) were fluorescence-activated cell sorted into MCAM (CD146)(+)/PDGFRB(+) (eMSC), MCAM (CD146)(-)/PDGFRB(+) (fibroblast), and MCAM (CD146)(+)/PDGFRB(-) (endothelial) populations. The eMSC population contained clonogenic cells with a mesenchymal phenotype differentiating into adipocytes when cultured in adipogenic medium. Gene expression profiling using Affymetrix Human Gene 1.0 ST arrays revealed 762 and 1518 significantly differentially expressed genes in eMSCs vs. stromal fibroblasts and eMSCs vs. endothelial cells, respectively. By principal component and hierarchical clustering analyses, eMSCs clustered with fibroblasts and distinctly from endothelial cells. Endometrial MSCs expressed pericyte markers and were localized by immunofluorescence to the perivascular space of endometrial small vessels. Endometrial MSCs also expressed genes involved in angiogenesis/vasculogenesis, steroid hormone/hypoxia responses, inflammation, immunomodulation, cell communication, and proteolysis/inhibition, and exhibited increased Notch, TGFB, IGF, Hedgehog, and G-protein-coupled receptor signaling pathways, characteristic of adult tissue MSC self-renewal and multipotency. Overall, the data support the eMSC as a clonogenic, multipotent pericyte that displays pathways of self-renewal and lineage specification, the potential to respond to conditions during endometrial desquamation and regeneration, and a genetic program predictive of its differentiated lineage, the stromal fibroblast.


Asunto(s)
Linaje de la Célula , Endometrio/fisiología , Células Madre Mesenquimatosas/fisiología , Pericitos/fisiología , Fenotipo , Regeneración/fisiología , Transducción de Señal/fisiología , Diferenciación Celular/fisiología , Células Cultivadas , Endometrio/citología , Endotelio/citología , Endotelio/fisiología , Femenino , Fibroblastos/citología , Fibroblastos/fisiología , Perfilación de la Expresión Génica , Humanos , Células Madre Mesenquimatosas/citología , Pericitos/citología , Transcriptoma/fisiología
13.
Fertil Steril ; 95(6): 2120-2, 2122.e1-12, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21371706

RESUMEN

OBJECTIVE: To develop a standard operating procedure (SOP) for collection, transport, storage of human endometrial tissue and blood samples, subject and specimen annotation, and establishing sample priorities. DESIGN: The SOP synthesizes sound scientific procedures, the literature on ischemia research, sample collection and gene expression profiling, good laboratory practices, and the authors' experience of workflow and sample quality. SETTING: The National Institutes of Health, University of California, San Francisco, Human Endometrial Tissue and DNA Bank. PATIENT(S): Women undergoing endometrial biopsy or hysterectomy for nonmalignant indications. INTERVENTION(S): Collecting, processing, storing, distributing endometrial tissue and blood samples under approved institutional review board protocols and written informed consent from participating subjects. MAIN OUTCOME MEASURE(S): Standard operating procedure. RESULT(S): The SOP addresses rigorous and consistent subject annotation, specimen processing and characterization, strict regulatory compliance, and a reference for researchers to track collection and storage times that may influence their research. CONCLUSION(S): The comprehensive and systematic approach to the procurement of human blood and endometrial tissue in this SOP ensures the high quality, reliability, and scientific usefulness of biospecimens made available to investigators by the National Institutes of Health, University of California, San Francisco, Human Endometrial Tissue and DNA Bank. The detail and perspective in this SOP also provides a blueprint for implementation of similar collection programs at other institutions.


Asunto(s)
Investigación Biomédica/métodos , Recolección de Muestras de Sangre/normas , Técnicas de Diagnóstico Obstétrico y Ginecológico/normas , Endometrio/cirugía , Procedimientos Quirúrgicos Ginecológicos/normas , Reproducción , Bancos de Tejidos/normas , Biología/métodos , Biología/normas , Investigación Biomédica/normas , Biopsia/métodos , Biopsia/normas , Recolección de Muestras de Sangre/métodos , Endometrio/patología , Femenino , Procedimientos Quirúrgicos Ginecológicos/métodos , Humanos , Etiquetado de Productos/normas , Control de Calidad , Estándares de Referencia , Reproducción/fisiología , Manejo de Especímenes/métodos , Manejo de Especímenes/normas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...