Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
ESMO Open ; 9(2): 102235, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38320429

RESUMEN

BACKGROUND: The use of circulating tumor DNA (ctDNA) concentration for metastatic cancer surveillance is promising, but uncertainty remains about cut-offs with clinical validity. MATERIALS AND METHODS: This observational study recruited 136 subjects with advanced metastatic breast cancer (irrespective of ERBB2/hormone receptor status) for sequencing of their primary tumor in search for PIK3CA hotspot variants amenable for monitoring by droplet digital PCR (ddPCR). The study analyzed 341 on-treatment samples from 19 patients with PIK3CA variants H1047R or E545K enrolled for long-term (median 85 weeks, range 13-125 weeks), frequent (every 3-5 weeks, median of 14 time points per subject, range 2-29) blood sampling for ctDNA quantification by ddPCR, orthogonally validated by deep sequencing. The diagnostic accuracy of ctDNA versus cancer antigen 15-3 (CA15-3) concentrations to predict disease progression within 12 weeks was investigated using receiver operating characteristic (ROC) analysis. Likelihood ratios were used for rational selection of ctDNA result intervals. RESULTS: ctDNA [area under the ROC curve (AUC) 0.848, 95% confidence interval (CI) 0.791-0.895] showed superior diagnostic performance than CA15-3 (AUC 0.670, 95% CI 0.601-0.735, P < 0.001) to predict clinical progression within 12 weeks. ctDNA levels below 10 mutant allele copies/ml had high negative predictive value (88%), while levels above 100 copies/ml detected 64% of progressions 10 weeks earlier versus standard of care. Logistic regression analysis indicated complementary value of ctDNA and the presence of two consecutive CA15-3 rises, resulting in a model with 86% (95% CI 74% to 93%) positive predictive value and a clinically meaningful result in 89% of blood draws. CONCLUSIONS: Intensive ctDNA quantification improves metastatic breast cancer surveillance and enables individualized risk-based scheduling of clinical care.


Asunto(s)
Neoplasias de la Mama , ADN Tumoral Circulante , Humanos , Femenino , ADN Tumoral Circulante/genética , Neoplasias de la Mama/tratamiento farmacológico , Biomarcadores de Tumor/genética , Progresión de la Enfermedad , Fosfatidilinositol 3-Quinasa Clase I/genética
2.
ESMO Open ; 6(5): 100274, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34597941

RESUMEN

BACKGROUND: Cancer patients are at a higher risk of developing severe coronavirus disease 2019 (COVID-19). However, the safety and efficacy of COVID-19 vaccination in cancer patients undergoing treatment remain unclear. PATIENTS AND METHODS: In this interventional prospective multicohort study, priming and booster doses of the BNT162b2 COVID-19 vaccine were administered 21 days apart to solid tumor patients receiving chemotherapy, immunotherapy, targeted or hormonal therapy, and patients with a hematologic malignancy receiving rituximab or after allogeneic hematopoietic stem cell transplantation. Vaccine safety and efficacy (until 3 months post-booster) were assessed. Anti-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) receptor-binding domain (RBD) antibody levels were followed over time (until 28 days after the booster) and in vitro SARS-CoV-2 50% neutralization titers (NT50) toward the wild-type Wuhan strain were analyzed 28 days after the booster. RESULTS: Local and systemic adverse events (AEs) were mostly mild to moderate (only 1%-3% of patients experienced severe AEs). Local, but not systemic, AEs occurred more frequently after the booster dose. Twenty-eight days after the booster vaccination of 197 cancer patients, RBD-binding antibody titers and NT50 were lower in the chemotherapy group {234.05 IU/ml [95% confidence interval (CI) 122.10-448.66] and 24.54 (95% CI 14.50-41.52), respectively} compared with healthy individuals [1844.93 IU/ml (95% CI 1383.57-2460.14) and 122.63 (95% CI 76.85-195.67), respectively], irrespective of timing of vaccination during chemotherapy cycles. Extremely low antibody responses were seen in hematology patients receiving rituximab; only two patients had RBD-binding antibody titers necessary for 50% protection against symptomatic SARS-CoV-2 infection (<200 IU/ml) and only one had NT50 above the limit of detection. During the study period, five cancer patients tested positive for SARS-CoV-2 infection, including a case of severe COVID-19 in a patient receiving rituximab, resulting in a 2-week hospital admission. CONCLUSION: The BNT162b2 vaccine is well-tolerated in cancer patients under active treatment. However, the antibody response of immunized cancer patients was delayed and diminished, mainly in patients receiving chemotherapy or rituximab, resulting in breakthrough infections.


Asunto(s)
Antineoplásicos , COVID-19 , Neoplasias , Vacuna BNT162 , Vacunas contra la COVID-19 , Humanos , Inmunidad Humoral , Estudios Prospectivos , ARN Mensajero , SARS-CoV-2 , Vacunación
3.
Cell Death Dis ; 7(6): e2272, 2016 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-27336716

RESUMEN

Partial pancreatic duct ligation (PDL) of mouse pancreas induces a doubling of the ß-cell mass mainly through proliferation of pre-existing and newly formed ß-cells. The molecular mechanism governing this process is still largely unknown. Given the inflammatory nature of PDL and inflammation-induced signaling via the signal transducer and activator of transcription 3 (STAT3), the activation and the role of STAT3 in PDL-induced ß-cell proliferation were investigated. Duct ligation stimulates the expression of several cytokines that can act as ligands inducing STAT3 signaling and phosphorylation in ß-cells. ß-Cell cycling increased by conditional ß-cell-specific Stat3 knockout and decreased by STAT3 activation through administration of interleukin-6. In addition, the level of DNA damage in ß-cells of PDL pancreas increased after deletion of Stat3. These data indicate a role for STAT3 in maintaining a steady state in the ß-cell, by modulating its cell cycle and protection from DNA damage.


Asunto(s)
Ciclo Celular , Citoprotección , Daño del ADN , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Factor de Transcripción STAT3/metabolismo , Animales , Ciclo Celular/efectos de los fármacos , Citocinas/metabolismo , Citoprotección/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Interleucina-6/farmacología , Antígeno Ki-67/metabolismo , Ligadura , Masculino , Ratones Endogámicos BALB C , Ratones Noqueados , Conductos Pancreáticos/efectos de los fármacos , Conductos Pancreáticos/patología , Proteínas Recombinantes/farmacología
4.
Data Brief ; 3: 234-9, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26217750

RESUMEN

Data set description: This data set is composed by label-free alternate-scanning LC-MS/MS proteomics analysis human and Wistar rat pancreatic islet endocrine cells. The mass spectrometry data of the human and rat pancreatic beta cells and the resulting proteome search output from ProteinLynx GlobalSERVER (PLGS) have been deposited to the ProteomeXchange Consortium [1] via the PRIDE partner repository with the dataset identifiers PXD001539 (human) and PXD001816 (rat). From these mass spectrometry data, 'relative molar amount units' between cell types and across species were calculated. Biological relevance: These data provide a quantitative view on the unfractionated proteomes of human and rat beta and alpha cells. It is likely biased towards the proteins with higher molar abundance, relating to core functional pathways, but also includes several proteins with an islet-enriched expression. The quality of the cell preps is state-of-the-art, and the label-free quantitation is both precise and accurate, allowing detailed quantitative analysis.

5.
J Diabetes Res ; 2015: 549818, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26064985

RESUMEN

The core proteomes of human and rat pancreatic beta cells were compared by label-free LC-MS/MS: this resulted in quantification of relative molar abundances of 707 proteins belonging to functional pathways of intermediary metabolism, protein synthesis, and cytoskeleton. Relative molar abundances were conserved both within and between pathways enabling the selection of a housekeeping network for geometric normalization and the analysis of potentially relevant differential expressions. Human beta cells differed from rat beta cells in their lower level of enzymes involved in glucose sensing (MDH1, PC, and ACLY) and upregulation of lysosomal enzymes. Human cells also expressed more heat shock proteins and radical scavenging systems: apart from SOD2, they expressed high levels of H2O2-scavenger peroxiredoxin 3 (PRDX3), confirmed by microarray, Western blotting, and microscopy. Besides conferring lower susceptibility to oxidative stress to human cells PRDX3 might also play a role in physiological redox regulation as, in rat, its expression was restricted to a beta cell subset with higher metabolic glucose responsiveness. In conclusion, although their core proteomic architecture is conserved, human and rat beta cells differ in their molar expression of key enzymes involved in glucose sensing and redox control.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Estrés Oxidativo/fisiología , Proteoma , Animales , Humanos , Proteómica , Ratas , Especificidad de la Especie , Espectrometría de Masas en Tándem , Regulación hacia Arriba
6.
J Proteomics ; 117: 156-67, 2015 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-25638021

RESUMEN

There is a clinical need for plasma tests for real-time detection of beta cell destruction, as surrogate endpoint in islet transplantation and immunoprevention trials in type 1 diabetes. This study reports on the use of label-free LC-MS/MS proteomics for bottom-up selection of candidate biomarkers. Ubiquitin COOH-terminal hydrolase 1 (UCHL1) was identified as abundant protein in rat and human beta cells, showing promising beta cell-selectivity, and was selected for further validation in standardized toxicity models. In vitro, H2O2-induced necrosis of INS-1 cells and human islets resulted in intracellular UCHL1 depletion and its extracellular discharge. In vivo, streptozotocin progressively depleted UCHL1 from islet cores and in 50% of animals, an associated plasma UCHL1 surge was detected preceding the GAD65 peak. UCHL1 was cleared with a half-life of 20min. Whole-body dynamic planar imaging of (99m)-Technetium-labeled UCHL1 indicated a rapid UCHL1 uptake in the liver and spleen, followed by urinary excretion of mainly proteolytic UCHL1 fragments. We conclude that LC-MS/MS proteomics is a useful tool to prioritize biomarkers for beta cell injury with promising molar abundance. Despite its consistent UCHL1 discharge by damaged beta cells in vitro, its in vivo use might be restrained by its rapid elimination from plasma. BIOLOGICAL SIGNIFICANCE: Our bottom-up LC-MS/MS proteomics represents a pragmatic approach to identify protein-type biomarkers of pancreatic beta cell injury. UCHL1 successfully passed sequential validation steps of beta cell-selectivity, antigenicity and toxic discharge in vitro. Whole-body dynamic planar imaging of radiolabeled recombinant UCHL1 indicated rapid clearance through the liver, spleen and urinary excretion of proteolytic fragments, likely explaining non-consistent detection in vivo. Integration of kinetic biomarker clearance studies in the a priori selection criteria is recommended before engaging in resource-intensive custom development of sensitive immunoassays for clinical translation.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Células Secretoras de Insulina/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Animales , Biomarcadores/metabolismo , Línea Celular , Diabetes Mellitus Experimental/patología , Humanos , Células Secretoras de Insulina/patología , Necrosis/metabolismo , Necrosis/patología , Ratas , Ubiquitina Tiolesterasa/genética
7.
J Mol Endocrinol ; 52(1): 11-28, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24049066

RESUMEN

Neonatal ß cells are considered developmentally immature and hence less glucose responsive. To study the acquisition of mature glucose responsiveness, we compared glucose-regulated redox state, insulin synthesis, and secretion of ß cells purified from neonatal or 10-week-old rats with their transcriptomes and proteomes measured by oligonucleotide and LC-MS/MS profiling. Lower glucose responsiveness of neonatal ß cells was explained by two distinct properties: higher activity at low glucose and lower activity at high glucose. Basal hyperactivity was associated with higher NAD(P)H, a higher fraction of neonatal ß cells actively incorporating (3)H-tyrosine, and persistently increased insulin secretion below 5 mM glucose. Neonatal ß cells lacked the steep glucose-responsive NAD(P)H rise between 5 and 10 mM glucose characteristic for adult ß cells and accumulated less NAD(P)H at high glucose. They had twofold lower expression of malate/aspartate-NADH shuttle and most glycolytic enzymes. Genome-wide profiling situated neonatal ß cells at a developmental crossroad: they showed advanced endocrine differentiation when specifically analyzed for their mRNA/protein level of classical neuroendocrine markers. On the other hand, discrete neonatal ß cell subpopulations still expressed mRNAs/proteins typical for developing/proliferating tissues. One example, delta-like 1 homolog (DLK1) was used to investigate whether neonatal ß cells with basal hyperactivity corresponded to a more immature subset with high DLK1, but no association was found. In conclusion, the current study supports the importance of glycolytic NADH-shuttling in stimulus function coupling, presents basal hyperactivity as novel property of neonatal ß cells, and provides potential markers to recognize intercellular developmental differences in the endocrine pancreas.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Animales , Animales Recién Nacidos , Biomarcadores/metabolismo , Diferenciación Celular , Análisis por Conglomerados , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Glucosa/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/citología , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Factor de Transcripción MafB/genética , Factor de Transcripción MafB/metabolismo , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Metabolómica , NAD/metabolismo , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Oxidación-Reducción , Proteómica , Ratas
8.
J Proteomics ; 80: 268-80, 2013 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-23337804

RESUMEN

There is a clinical need for plasma tests that can directly detect injury to pancreatic beta cells in type 1 diabetes. Such tests require biomarkers that are abundantly and selectively released into plasma by damaged beta cells. We combined LC-MS/MS proteomics and tissue-comparative transcriptomics of FACS-purified beta cells for bottom-up identification of candidate markers. Less than 10% of 467 proteins detected in beta cells showed endocrine-enriched expression. One surprising candidate was the neuronal migration marker doublecortin: in situ analysis revealed uniform doublecortin expression in the cytoplasm of all beta cells. Western blotting and real-time PCR confirmed its strong beta cell-selectivity outside the brain and its high molar abundance, indicating promising biomarker properties in comparison to GAD65, a more established marker of beta cell injury. DCX potential was validated in vitro: chemically-induced necrosis of rat and human beta cells led to a discharge of intracellular doublecortin into the extracellular space, proportionate to the amount of injured cells, and similar to GAD65. In vivo, recombinant DCX showed favorable pharmacokinetic properties, with a half-life in plasma of around 3h. Combined, our findings provide first proof-of-principle for doublecortin as biomarker for beta cell injury in vitro, advocating its further validation as biomarker in vivo.


Asunto(s)
Biomarcadores/análisis , Células Secretoras de Insulina/patología , Proteínas Asociadas a Microtúbulos/metabolismo , Neuropéptidos/metabolismo , Animales , Proteínas de Dominio Doblecortina , Proteína Doblecortina , Glutamato Descarboxilasa/sangre , Humanos , Células Secretoras de Insulina/metabolismo , Proteínas Asociadas a Microtúbulos/inmunología , Proteínas Asociadas a Microtúbulos/aislamiento & purificación , Neuropéptidos/inmunología , Neuropéptidos/aislamiento & purificación , Proteómica , Ratas , Transcriptoma
9.
Diabetologia ; 54(6): 1273-90, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21442161

RESUMEN

The prevalence of diabetes mellitus is currently at epidemic proportions and it is estimated that it will increase even further over the next decades. Although genetic predisposition and lifestyle choices are commonly accepted reasons for the occurrence of type 2 diabetes, it has recently been suggested that environmental pollutants are additional risk factors for diabetes development and this review aims to give an overview of the current evidence for this. More specifically, because of the crucial role of pancreatic beta cells in the development and progression of type 2 diabetes, the present work summarises the known effects of several compounds on beta cell function with reference to mechanistic studies that have elucidated how these compounds interfere with the insulin secreting capacity of beta cells. Oestrogenic compounds, organophosphorus compounds, persistent organic pollutants and heavy metals are discussed, and a critical reflection on the relevance of the concentrations used in mechanistic studies relative to the levels found in the human population is given. It is clear that some environmental pollutants affect pancreatic beta cell function, as both epidemiological and experimental research is accumulating. This supports the need to develop a solid and structured platform to fully explore the diabetes-inducing potential of pollutants.


Asunto(s)
Diabetes Mellitus Tipo 2/epidemiología , Contaminantes Ambientales/efectos adversos , Células Secretoras de Insulina/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Contaminantes Ambientales/farmacología , Estrógenos/efectos adversos , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Metales Pesados/efectos adversos , Compuestos Organofosforados/efectos adversos , Factores de Riesgo
10.
Br J Pharmacol ; 150(8): 1031-43, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17339833

RESUMEN

BACKGROUND AND PURPOSE: Two mechanisms have been proposed to explain the insulin-sensitising properties of metformin in peripheral tissues: (a) inhibition of electron transport chain complex I, and (b) activation of the AMP activated protein kinase (AMPK). However the relationship between these mechanisms and their contribution to beta-cell death and dysfunction in vitro, are currently unclear. EXPERIMENTAL APPROACH: The effects of biguanides (metformin and phenformin) were tested on MIN6 beta-cells and primary FACS-purified rat beta-cells. Cell metabolism was assessed biochemically and by FACS analysis, and correlated with AMPK phosphorylation state and cell viability, with or without fuel substrates. KEY RESULTS: In MIN6 cells, metformin reduced mitochondrial complex I activity by up to 44% and a 25% net reduction in mitochondrial reducing potential. In rat beta-cells, metformin caused NAD(P)H accumulation above maximal glucose-inducible levels, mimicking the effect of rotenone. Drug exposure caused phosphorylation of AMPK on Thr(172) in MIN6 cell extracts, indicative of kinase activation. Methyl succinate, a complex II substrate, appeared to bypass metformin blockade of complex I. This resulted in reduced phosphorylation of AMPK, establishing a link between biguanide-induced mitochondrial inhibition and AMPK activation. Corresponding assessment of cell death indicated that methyl succinate decreased biguanide toxicity to beta-cells in vitro. CONCLUSIONS AND IMPLICATIONS: AMPK activation can partly be attributed to metformin's inhibitory action on mitochondrial complex I. Anaplerotic fuel metabolism via complex II rescued beta-cells from metformin-associated toxicity. We propose that utilisation of anaplerotic nutrients may reconcile in vitro and in vivo effects of metformin on the pancreatic beta-cell.


Asunto(s)
Biguanidas/toxicidad , Hipoglucemiantes/toxicidad , Células Secretoras de Insulina/efectos de los fármacos , Complejos Multienzimáticos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Succinatos/farmacología , Proteínas Quinasas Activadas por AMP , Animales , Apoptosis/efectos de los fármacos , Biguanidas/antagonistas & inhibidores , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Transporte de Electrón/efectos de los fármacos , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Complejo I de Transporte de Electrón/metabolismo , Complejo II de Transporte de Electrones/efectos de los fármacos , Complejo II de Transporte de Electrones/metabolismo , Activación Enzimática/efectos de los fármacos , Glucosa/metabolismo , Hipoglucemiantes/antagonistas & inhibidores , Células Secretoras de Insulina/metabolismo , Metformina/toxicidad , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , NADP/metabolismo , Oxidación-Reducción , Fenformina/toxicidad , Fosforilación/efectos de los fármacos , Ratas , Succinatos/metabolismo , Sales de Tetrazolio/metabolismo , Tiazoles/metabolismo , Factores de Tiempo
11.
Endocrinology ; 147(11): 5196-204, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16916947

RESUMEN

Insulin synthesis and release activities of beta-cells can be acutely regulated by glucose through its glycolytic and mitochondrial breakdown involving a glucokinase-dependent rate-limiting step. Isolated beta-cell populations are composed of cells with intercellular differences in acute glucose responsiveness that have been attributed to differences in glucokinase (GK) expression and activity. This study first shows that glyceraldehyde can be used as GK-bypassing oxidative substrate and then examines whether the triose can metabolically activate beta-cells with low glucose responsiveness. Glyceraldehyde 1 mm induced a similar cellular (14)CO(2) output and metabolic redox state as glucose 4 mM. Using flow cytometric analysis, glyceraldehyde (0.25-2 mM) was shown to concentration-dependently increase the percent metabolically activated cells at all tested glucose concentrations (2.5-20 mM). Its ability to activate beta-cells that are unresponsive to the prevailing glucose level was further illustrated in glucose low-responsive cells that were isolated by flow sorting. Metabolic activation by glyceraldehyde was associated with an activation of nutrient-driven translational control proteins and an increased protein synthetic response to glucose, however not beyond the maximal rates that are inducible by glucose alone. It is concluded that glucose low-responsive beta-cells can be metabolically activated by the GK-bypassing glyceraldehyde, increasing their acute biosynthetic response to glucose but not their maximal glucose-inducible biosynthetic capacity, which is considered subject to chronic regulation.


Asunto(s)
Glucosa/farmacología , Gliceraldehído/farmacología , Islotes Pancreáticos/metabolismo , Biosíntesis de Proteínas/efectos de los fármacos , Animales , Glucoquinasa/fisiología , Glucosa/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Masculino , Oxidación-Reducción , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...