Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 23(15)2022 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-35897659

RESUMEN

Hepatocellular carcinoma (HCC) incidence, as well as related mortality, has been steadily increasing in the USA and across the globe, partly due to the lack of effective therapeutic options for advanced HCC. Though sorafenib is considered standard-of-care for advanced HCC, it only improves median survival by a few months when compared to placebo. Sorafenib is also associated with several unpleasant side effects that often lead to early abatement of therapy. Here, we investigate whether a combination regimen including low-dose sorafenib and a non-toxic dose of anti-diabetic drug metformin can achieve effective inhibition of HCC. Indeed, combining metformin with low-dose sorafenib inhibited growth, proliferation, migration, and invasion potential of HCC cells. We observed a 5.3- and 1.9-fold increase in sub-G1 population in the combination treatment compared to sorafenib alone. We found that the combination of metformin enhanced the efficacy of sorafenib and inhibited the MAPK/ERK/Stat3 axis. Our in vivo studies corroborated the in vitro findings, and mice harboring HepG2-derived tumors showed effective tumor reduction upon treatment with low-dose sorafenib and metformin combination. This work sheds light on a therapeutic strategy aiming to augment sorafenib efficacy or dose-de-escalation that may prove beneficial in circumventing sorafenib resistance as well as minimizing related side effects.


Asunto(s)
Antineoplásicos , Carcinoma Hepatocelular , Neoplasias Hepáticas , Metformina , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Proliferación Celular , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Metformina/farmacología , Metformina/uso terapéutico , Ratones , Niacinamida/farmacología , Niacinamida/uso terapéutico , Compuestos de Fenilurea/farmacología , Compuestos de Fenilurea/uso terapéutico , Sorafenib/farmacología , Sorafenib/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Neuroimage ; 245: 118659, 2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34767940

RESUMEN

Studying changes in cortical oscillations can help elucidate the mechanistic link between receptor physiology and the clinical effects of anaesthetic drugs. Propofol, a GABA-ergic drug produces divergent effects on visual cortical activity: increasing induced gamma-band responses (GBR) while decreasing evoked responses. Dexmedetomidine, an α2- adrenergic agonist, differs from GABA-ergic sedatives both mechanistically and clinically as it allows easy arousability from deep sedation with less cognitive side-effects. Here we use magnetoencephalography (MEG) to characterize and compare the effects of GABA-ergic (propofol) and non-GABA-ergic (dexmedetomidine) sedation, on visual and motor cortical oscillations. Sixteen male participants received target-controlled infusions of propofol and dexmedetomidine, producing mild-sedation, in a placebo-controlled, cross-over study. MEG data was collected during a combined visuomotor task. The key findings were that propofol significantly enhanced visual stimulus induced GBR (44% increase in amplitude) while dexmedetomidine decreased it (40%). Propofol also decreased the amplitudes of the Mv100 (visual M100) (27%) and Mv150 (52%) visual evoked fields (VEF), whilst dexmedetomidine had no effect on these. During the motor task, neither drug had any significant effect on movement related gamma synchrony (MRGS), movement related beta de-synchronisation (MRBD) or Mm100 (movement-related M100) movement-related evoked fields (MEF), although dexmedetomidine slowed the Mm300. Dexmedetomidine increased (92%) post-movement beta synchronisation/rebound (PMBR) power while propofol reduced it (70%, statistically non- significant). Overall, dexmedetomidine and propofol, at equi-sedative doses, produce contrasting effects on visual induced GBR, VEF, PMBR and MEF. These findings provide a mechanistic link between the known receptor physiology of these sedative drugs with their known clinical effects and may be used to explore mechanisms of other anaesthetic drugs on human consciousness.


Asunto(s)
Ondas Encefálicas/efectos de los fármacos , Dexmedetomidina/farmacología , Hipnóticos y Sedantes/farmacología , Magnetoencefalografía/métodos , Corteza Motora/efectos de los fármacos , Propofol/farmacología , Adulto , Sedación Consciente , Estado de Conciencia/efectos de los fármacos , Estudios Cruzados , Humanos , Masculino , Movimiento/fisiología , Vigilia , Adulto Joven
3.
Neuroimage ; 221: 117189, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32711064

RESUMEN

Cortical recordings of task-induced oscillations following subanaesthetic ketamine administration demonstrate alterations in amplitude, including increases at high-frequencies (gamma) and reductions at low frequencies (theta, alpha). To investigate the population-level interactions underlying these changes, we implemented a thalamo-cortical model (TCM) capable of recapitulating broadband spectral responses. Compared with an existing cortex-only 4-population model, Bayesian Model Selection preferred the TCM. The model was able to accurately and significantly recapitulate ketamine-induced reductions in alpha amplitude and increases in gamma amplitude. Parameter analysis revealed no change in receptor time-constants but significant increases in select synaptic connectivity with ketamine. Significantly increased connections included both AMPA and NMDA mediated connections from layer 2/3 superficial pyramidal cells to inhibitory interneurons and both GABAA and NMDA mediated within-population gain control of layer 5 pyramidal cells. These results support the use of extended generative models for explaining oscillatory data and provide in silico support for ketamine's ability to alter local coupling mediated by NMDA, AMPA and GABA-A.


Asunto(s)
Ondas Encefálicas , Corteza Cerebral , Antagonistas de Aminoácidos Excitadores/farmacología , Interneuronas , Ketamina/farmacología , Magnetoencefalografía , Modelos Biológicos , Células Piramidales , Tálamo , Adolescente , Adulto , Ondas Encefálicas/efectos de los fármacos , Ondas Encefálicas/fisiología , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/fisiología , Humanos , Interneuronas/efectos de los fármacos , Interneuronas/fisiología , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Reconocimiento Visual de Modelos/efectos de los fármacos , Reconocimiento Visual de Modelos/fisiología , Células Piramidales/efectos de los fármacos , Células Piramidales/fisiología , Tálamo/efectos de los fármacos , Tálamo/fisiología , Adulto Joven
4.
Carcinogenesis ; 40(9): 1110-1120, 2019 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-30698683

RESUMEN

Withaferin A (WFA), a steroidal lactone, negatively regulates breast cancer growth however, its mechanisms of action remain largely elusive. We found that WFA blocks autophagy flux and lysosomal proteolytic activity in breast cancer cells. WFA increases accumulation of autophagosomes, LC3B-II conversion, expression of autophagy-related proteins and autophagosome/lysosome fusion. Autolysosomes display the characteristics of acidic compartments in WFA-treated cells; however, the protein degradation activity of lysosomes is inhibited. Blockade of autophagic flux reduces the recycling of cellular fuels leading to insufficient substrates for tricarboxylic acid (TCA) cycle and impaired oxidative phosphorylation. WFA decreases expression and phosphorylation of lactate dehydrogenase, the key enzyme that catalyzes pyruvate-to-lactate conversion, reduces adenosine triphosphate levels and increases AMP-activated protein kinase (AMPK) activation. AMPK inhibition abrogates while AMPK activation potentiates WFA's effect. WFA and 2-deoxy-d-glucose combination elicits synergistic inhibition of breast cancer cells. Genetic knockout of BECN1 and ATG7 fails to rescue cells from WFA treatment; in contrast, addition of methyl pyruvate to supplement TCA cycle protects WFA-treated cells. Together, these results implicate that WFA is a potent lysosomal inhibitor; energetic impairment is required for WFA-induced apoptosis and growth inhibition and combining WFA and 2-DG is a promising therapeutic strategy for breast cancer.

5.
Biochem J ; 474(5): 647-665, 2017 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-28008134

RESUMEN

M3 muscarinic receptor (M3R) expression is increased in colon cancer; M3R activation stimulates colon cancer cell invasion via cross-talk with epidermal growth factor receptors (EGFR), post-EGFR activation of mitogen-activated protein kinase (MAPK) extracellular signal-related kinase 1/2 (ERK1/2), and induction of matrix metalloproteinase-1 (MMP1) expression. MMP1 expression is strongly associated with tumor metastasis and adverse outcomes. Here, we asked whether other MAPKs regulate M3R agonist-induced MMP1 expression. In addition to activating ERK1/2, we found that treating colon cancer cells with acetylcholine (ACh) stimulated robust time- and dose-dependent phosphorylation of p38 MAPK. Unlike ERK1/2 activation, ACh-induced p38 phosphorylation was EGFR-independent and blocked by inhibiting protein kinase C-α (PKC-α). Inhibiting activation of PKC-α, EGFR, ERK1/2, or p38-α/ß alone attenuated, but did not abolish ACh-induced MMP1 expression, a finding that predicted potentiating interactions between these pathways. Indeed, ACh-induced MMP1 expression was abolished by incubating cells with either an EGFR or MEK/ERK1/2 inhibitor combined with a p38-α/ß inhibitor. Activating PKC-α and EGFR directly with the combination of phorbol 12-myristate 13-acetate (PMA) and EGF potentiated MMP1 gene and protein expression, and cell invasion. PMA- and ACh-induced MMP1 expression were strongly diminished by inhibiting Src and abolished by concurrently inhibiting both p38-α/ß and Src, indicating that Src mediates the cross-talk between PKC-α and EGFR signaling. Using siRNA knockdown, we identified p38-α as the relevant p38 isoform. Collectively, these studies uncover novel functional interactions between post-muscarinic receptor signaling pathways that augment MMP1 expression and drive colon cancer cell invasion; targeting these potentiating interactions has therapeutic potential.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Metaloproteinasa 1 de la Matriz/metabolismo , Receptor Muscarínico M3/genética , Transducción de Señal/genética , Acetilcolina/farmacología , Células CACO-2 , Línea Celular Tumoral , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/genética , Receptores ErbB/metabolismo , Células HT29 , Humanos , Metaloproteinasa 1 de la Matriz/genética , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación/efectos de los fármacos , Proteína Quinasa C-alfa/genética , Proteína Quinasa C-alfa/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptor Muscarínico M3/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
6.
Semin Cancer Biol ; 35 Suppl: S25-S54, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25892662

RESUMEN

Proliferation is an important part of cancer development and progression. This is manifest by altered expression and/or activity of cell cycle related proteins. Constitutive activation of many signal transduction pathways also stimulates cell growth. Early steps in tumor development are associated with a fibrogenic response and the development of a hypoxic environment which favors the survival and proliferation of cancer stem cells. Part of the survival strategy of cancer stem cells may manifested by alterations in cell metabolism. Once tumors appear, growth and metastasis may be supported by overproduction of appropriate hormones (in hormonally dependent cancers), by promoting angiogenesis, by undergoing epithelial to mesenchymal transition, by triggering autophagy, and by taking cues from surrounding stromal cells. A number of natural compounds (e.g., curcumin, resveratrol, indole-3-carbinol, brassinin, sulforaphane, epigallocatechin-3-gallate, genistein, ellagitannins, lycopene and quercetin) have been found to inhibit one or more pathways that contribute to proliferation (e.g., hypoxia inducible factor 1, nuclear factor kappa B, phosphoinositide 3 kinase/Akt, insulin-like growth factor receptor 1, Wnt, cell cycle associated proteins, as well as androgen and estrogen receptor signaling). These data, in combination with bioinformatics analyses, will be very important for identifying signaling pathways and molecular targets that may provide early diagnostic markers and/or critical targets for the development of new drugs or drug combinations that block tumor formation and progression.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proliferación Celular/efectos de los fármacos , Neoplasias/patología , Neoplasias/terapia , Antineoplásicos/uso terapéutico , Proteínas de Ciclo Celular/biosíntesis , Transición Epitelial-Mesenquimal/efectos de los fármacos , Humanos , Terapia Molecular Dirigida , Neoplasias/genética , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Transducción de Señal/efectos de los fármacos
7.
J Neurosci ; 35(33): 11694-706, 2015 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-26290246

RESUMEN

Following the discovery of the antidepressant properties of ketamine, there has been a recent resurgence in the interest in this NMDA receptor antagonist. Although detailed animal models of the molecular mechanisms underlying ketamine's effects have emerged, there are few MEG/EEG studies examining the acute subanesthetic effects of ketamine infusion in man. We recorded 275 channel MEG in two experiments (n = 25 human males) examining the effects of subanesthetic ketamine infusion. MEG power spectra revealed a rich set of significant oscillatory changes compared with placebo sessions, including decreases in occipital, parietal, and anterior cingulate alpha power, increases in medial frontal theta power, and increases in parietal and cingulate cortex high gamma power. Each of these spectral effects demonstrated their own set of temporal dynamics. Dynamic causal modeling of frontoparietal connectivity changes with ketamine indicated a decrease in NMDA and AMPA-mediated frontal-to-parietal connectivity. AMPA-mediated connectivity changes were sustained for up to 50 min after ketamine infusion had ceased, by which time perceptual distortions were absent. The results also indicated a decrease in gain of parietal pyramidal cells, which was correlated with participants' self-reports of blissful state. Based on these results, we suggest that the antidepressant effects of ketamine may depend on its ability to change the balance of frontoparietal connectivity patterns. SIGNIFICANCE STATEMENT: In this paper, we found that subanesthetic doses of ketamine, similar to those used in antidepressant studies, increase anterior theta and gamma power but decrease posterior theta, delta, and alpha power, as revealed by magnetoencephalographic recordings. Dynamic causal modeling of frontoparietal connectivity changes with ketamine indicated a decrease in NMDA and AMPA-mediated frontal-to-parietal connectivity. AMPA-mediated connectivity changes were sustained for up to 50 min after ketamine infusion had ceased, by which time perceptual distortions were absent. The results also indicated a decrease in gain of parietal pyramidal cells, which was correlated with participants' self-reports of blissful state. The alterations in frontoparietal connectivity patterns we observe here may be important in generating the antidepressant response to ketamine.


Asunto(s)
Ondas Encefálicas/fisiología , Lóbulo Frontal/fisiología , Ketamina/administración & dosificación , Lóbulo Parietal/fisiología , Receptores AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Adulto , Anestésicos Disociativos/administración & dosificación , Antidepresivos/administración & dosificación , Mapeo Encefálico , Ondas Encefálicas/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Lóbulo Frontal/efectos de los fármacos , Humanos , Masculino , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/fisiología , Lóbulo Parietal/efectos de los fármacos , Receptores AMPA/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Adulto Joven
8.
FASEB J ; 28(12): 5172-83, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25154876

RESUMEN

Previous evidence indicates that adiponectin possesses antifibrogenic activity in inhibiting liver fibrosis. Therapeutic strategies, however, are limited by adiponectin quaternary structure and effective concentrations in circulation. Here we postulate a novel molecular mechanism, whereby adiponectin targets focal adhesion kinase (FAK) activity and disrupts key features of the fibrogenic response. Adiponectin-null (Ad(-/-)) mice and wild-type littermates were exposed to either saline or carbon tetrachloride (CCl4) for 6 wk. CCl4-gavaged mice were also injected with attenuated adenoviral adiponectin (Ad-Adn) or Ad-LacZ for 2 wk. Hepatic stellate cells (HSCs) were treated with or without adiponectin to elucidate signal transduction mechanisms. In vivo delivery of Ad-Adn markedly attenuates CCl4-induced expression of key integrin proteins and markers of HSC activation: αv, ß3, ß1, α2(I) collagen, and α-smooth muscle actin. Confocal experiments of liver tissues demonstrated that adiponectin delivery also suppressed vinculin and p-FAK activity in activated HSCs. In vitro, adiponectin induced dephosphorylation of FAK, mediated by a physical association with activated tyrosine phosphatase, Shp2. Conversely, Shp2 knockdown by siRNA significantly attenuated adiponectin-induced FAK deactivation, and expression of TIMP1 and α2(I) collagen was abolished in the presence of adiponectin and si-FAK. Finally, we documented that either adiponectin or the synthetic peptide with adiponectin properties, ADP355, suppressed p-FAK in synthetic matrices with stiffness measurements of 9 and 15 kPa, assessed by immunofluorescent imaging and quantitation. The in vivo and in vitro data presented indicate that disassembly of focal adhesion complexes in HSCs is pivotal for hepatic fibrosis therapy, now that small adiponectin-like peptides are available.


Asunto(s)
Adiponectina/fisiología , Adhesiones Focales , Células Estrelladas Hepáticas/citología , Cirrosis Hepática/terapia , Animales , Secuencia de Bases , Cartilla de ADN , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
9.
J Neurosci ; 33(9): 4024-31, 2013 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-23447611

RESUMEN

Despite their routine use during surgical procedures, no consensus has yet been reached on the precise mechanisms by which hypnotic anesthetic agents produce their effects. Molecular, animal and human studies have suggested disruption of thalamocortical communication as a key component of anesthetic action at the brain systems level. Here, we used the anesthetic agent, propofol, to modulate consciousness and to evaluate differences in the interactions of remote neural networks during altered consciousness. We investigated the effects of propofol, at a dose that produced mild sedation without loss of consciousness, on spontaneous cerebral activity of 15 healthy volunteers using functional magnetic resonance imaging (fMRI), exploiting oscillations (<0.1 Hz) in blood oxygenation level-dependent signal across functionally connected brain regions. We considered the data as a graph, or complex network of nodes and links, and used eigenvector centrality (EC) to characterize brain network properties. The EC mapping of fMRI data in healthy humans under propofol mild sedation demonstrated a decrease of centrality of the thalamus versus an increase of centrality within the pons of the brainstem, highlighting the important role of these two structures in regulating consciousness. Specifically, the decrease of thalamus centrality results from its disconnection from a widespread set of cortical and subcortical regions, while the increase of brainstem centrality may be a consequence of its increased influence, in the mildly sedated state, over a few highly central cortical regions key to the default mode network such as the posterior and anterior cingulate cortices.


Asunto(s)
Anestésicos Intravenosos/farmacología , Mapeo Encefálico , Tronco Encefálico/efectos de los fármacos , Vías Nerviosas/fisiología , Propofol/farmacología , Tálamo/efectos de los fármacos , Adulto , Tronco Encefálico/irrigación sanguínea , Humanos , Procesamiento de Imagen Asistido por Computador , Imagen por Resonancia Magnética , Masculino , Red Nerviosa/irrigación sanguínea , Red Nerviosa/efectos de los fármacos , Vías Nerviosas/irrigación sanguínea , Vías Nerviosas/efectos de los fármacos , Oxígeno/sangre , Tálamo/irrigación sanguínea , Vigilia , Adulto Joven
10.
J Mammary Gland Biol Neoplasia ; 18(3-4): 309-20, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24214584

RESUMEN

High plasma levels of leptin, a major adipocytokine produced by adipocytes, are correlated with increased fat mass in obese state. Leptin is emerging as a key candidate molecule linking obesity with breast cancer. Acting via endocrine, paracrine, and autocrine manner, leptin impacts various stages of breast tumorigenesis from initiation and primary tumor growth to metastatic progression. Leptin also modulates the tumor microenvironment mainly through supporting migration of endothelial cells, neo-angiogenesis and sustaining recruitment of macrophage and monocytes. Various studies have shown that hyperactive leptin-signaling network leads to concurrent activation of multiple oncogenic pathways resulting in enhanced proliferation, decreased apoptosis, acquisition of mesenchymal phenotype, potentiated migration and enhanced invasion potential of tumor cells. Furthermore, the capability of leptin to interact with other molecular effectors of obese state including, estrogen, IGF-1, insulin, VEGF and inflammatory cytokines further increases its impact on breast tumor progression in obese state. This article presents an overview of the studies investigating the involvement of leptin in breast cancer.


Asunto(s)
Neoplasias de la Mama/fisiopatología , Leptina/fisiología , Obesidad/fisiopatología , Animales , Neoplasias de la Mama/metabolismo , Femenino , Humanos , Obesidad/metabolismo , Receptores de Leptina/metabolismo , Factores de Riesgo , Transducción de Señal
11.
J Biol Chem ; 287(11): 8598-612, 2012 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-22270359

RESUMEN

Perturbations in the adipocytokine profile, especially higher levels of leptin, are a major cause of breast tumor progression and metastasis; the underlying mechanisms, however, are not well understood. In particular, it remains elusive whether leptin is involved in epithelial-mesenchymal transition (EMT). Here, we provide molecular evidence that leptin induces breast cancer cells to undergo a transition from epithelial to spindle-like mesenchymal morphology. Investigating the downstream mediator(s) that may direct leptin-induced EMT, we found functional interactions between leptin, metastasis-associated protein 1 (MTA1), and Wnt1 signaling components. Leptin increases accumulation and nuclear translocation of ß-catenin leading to increased promoter recruitment. Silencing of ß-catenin or treatment with the small molecule inhibitor, ICG-001, inhibits leptin-induced EMT, invasion, and tumorsphere formation. Mechanistically, leptin stimulates phosphorylation of glycogen synthase kinase 3ß (GSK3ß) via Akt activation resulting in a substantial decrease in the formation of the GSK3ß-LKB1-Axin complex that leads to increased accumulation of ß-catenin. Leptin treatment also increases Wnt1 expression that contributes to GSK3ß phosphorylation. Inhibition of Wnt1 abrogates leptin-stimulated GSK3ß phosphorylation. We also discovered that leptin increases the expression of an important modifier of Wnt1 signaling, MTA1, which is integral to leptin-mediated regulation of the Wnt/ß-catenin pathway as silencing of MTA1 inhibits leptin-induced Wnt1 expression, GSK3ß phosphorylation, and ß-catenin activation. Furthermore, analysis of leptin-treated breast tumors shows increased expression of Wnt1, pGSK3ß, and vimentin along with higher nuclear accumulation of ß-catenin and reduced E-cadherin expression providing in vivo evidence for a previously unrecognized cross-talk between leptin and MTA1/Wnt signaling in epithelial-mesenchymal transition of breast cancer cells.


Asunto(s)
Neoplasias de la Mama/metabolismo , Transición Epitelial-Mesenquimal , Glucógeno Sintasa Quinasa 3/metabolismo , Histona Desacetilasas/metabolismo , Leptina/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Represoras/metabolismo , Vía de Señalización Wnt , Proteína Wnt1/metabolismo , beta Catenina/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Cadherinas/genética , Cadherinas/metabolismo , Línea Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Núcleo Celular/patología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Histona Desacetilasas/genética , Humanos , Leptina/genética , Leptina/farmacología , Fosforilación/efectos de los fármacos , Fosforilación/genética , Proteínas Proto-Oncogénicas c-akt/genética , Pirimidinonas/farmacología , Proteínas Represoras/genética , Transactivadores , Vimentina/genética , Vimentina/metabolismo , Proteína Wnt1/genética , beta Catenina/antagonistas & inhibidores , beta Catenina/genética
12.
NMR Biomed ; 26(12): 1844-52, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24123253

RESUMEN

Hypoxic hypoxia (inspiratory hypoxia) stimulates an increase in cerebral blood flow (CBF) maintaining oxygen delivery to the brain. However, this response, particularly at the tissue level, is not well characterised. This study quantifies the CBF response to acute hypoxic hypoxia in healthy subjects. A 20-min hypoxic (mean P(ETO2) = 52 mmHg) challenge was induced and controlled by dynamic end-tidal forcing whilst CBF was measured using pulsed arterial spin labelling perfusion MRI. The rate constant, temporal delay and magnitude of the CBF response were characterised using an exponential model for whole-brain and regional grey matter. Grey matter CBF increased from 76.1 mL/100 g/min (95% confidence interval (CI) of fitting: 75.5 mL/100 g/min, 76.7 mL/100 g/min) to 87.8 mL/100 g/min (95% CI: 86.7 mL/100 g/min, 89.6 mL/100 g/min) during hypoxia, and the temporal delay and rate constant for the response to hypoxia were 185 s (95% CI: 132 s, 230 s) and 0.0035 s(-1) (95% CI: 0.0019 s(-1), 0.0046 s(-1)), respectively. Recovery from hypoxia was faster with a delay of 20 s (95% CI: -38 s, 38 s) and a rate constant of 0.0069 s(-1) (95% CI: 0.0020 s(-1), 0.0103 s(-1)). R2*, an index of blood oxygenation obtained simultaneously with the CBF measurement, increased from 30.33 s(-1) (CI: 30.31 s(-1), 30.34 s(-1)) to 31.48 s(-1) (CI: 31.47 s(-1), 31.49 s(-1)) with hypoxia. The delay and rate constant for changes in R2 * were 24 s (95% CI: 21 s, 26 s) and 0.0392 s(-1) (95% CI: 0.0333 s(-1), 0.045 s(-1)), respectively, for the hypoxic response, and 12 s (95% CI: 10 s, 13 s) and 0.0921 s(-1) (95% CI: 0.0744 s(-1), 0.1098 s(-1)/) during the return to normoxia, confirming rapid changes in blood oxygenation with the end-tidal forcing system. CBF and R2* reactivity to hypoxia differed between subjects, but only R2* reactivity to hypoxia differed significantly between brain regions.


Asunto(s)
Circulación Cerebrovascular/fisiología , Hipoxia/fisiopatología , Enfermedad Aguda , Adulto , Femenino , Humanos , Masculino , Modelos Biológicos
13.
J Magn Reson Imaging ; 37(3): 739-45, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23197421

RESUMEN

PURPOSE: To demonstrate the feasibility of measuring the temporal dynamics of cerebral lactate concentration and examine these dynamics in human subjects using magnetic resonance spectroscopy (MRS) during hypoxia. MATERIALS AND METHODS: A respiratory protocol consisting of 10-minute baseline normoxia, 20-minute inspiratory hypoxia, and ending with 10-minute normoxic recovery was used, throughout which lactate-edited MRS was performed. This was repeated four times in three subjects. A separate session was performed to measure blood lactate. Impulse response functions using end-tidal oxygen and blood lactate as system inputs and cerebral lactate as the system output were examined to describe the dynamics of the cerebral lactate response to a hypoxic challenge. RESULTS: The average lactate increase was 20% ± 15% during the last half of the hypoxic challenge. Significant changes in cerebral lactate concentration were observed after 400 seconds. The average relative increase in blood lactate was 188% ± 95%. The temporal dynamics of cerebral lactate concentration was reproducibly demonstrated with 200-second time bins of MRS data (coefficient of variation 0.063 ± 0.035 between time bins in normoxia). The across-subject coefficient of variation was 0.333. CONCLUSION: The methods for measuring the dynamics of the cerebral lactate response developed here would be useful to further investigate the brain's response to hypoxia.


Asunto(s)
Encéfalo/patología , Hipoxia Encefálica/patología , Lactatos/metabolismo , Espectroscopía de Resonancia Magnética/métodos , Enfermedad Aguda , Adulto , Femenino , Humanos , Hipoxia , Lactatos/sangre , Ácido Láctico/metabolismo , Masculino , Modelos Estadísticos , Oxígeno/química , Respiración , Factores de Tiempo
14.
Pol Przegl Chir ; 96(0): 123-129, 2023 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-38348981

RESUMEN

<b><br>Introduction:</b> Wound infection is the most common post-operative complication encountered after open appendectomy. Various studies have compared the risk of superficial surgical site infection (SSI) in primary closure (PC) and delayed primary closure (DPC) of wounds. However, there is no uniform consensus regarding the method of wound closure.</br> <b><br>Aim:</b> The aim of this study is to compare the two wound closure techniques.</br> <b><br>Material and methods:</b> This is a prospective study which enrolled 50 patients who underwent open appendectomy. The patients' demographics, characteristics, and operative findings were recorded. Those who were older than 18 years and had an appendectomy with a right lower quadrant incision were included. Patients with any comorbidity, morbid obesity, or pregnancy were excluded. Patients were randomized to undergo two techniques of wound closure: PC and DPC. During follow- -up at 1 week and 1 month, SSI, post-op pain, and LOS were compared among the two groups. Clinical assessment included the Visual Analog Scale (1-10) for pain.</br> <b><br>Results:</b> In our study, the incidence of SSI in the DPC group was significantly lower than in the PC group (p = 0.0002), while post-op pain and LOS were not significantly different between the two groups.</br> <b><br>Conclusions:</b> We concluded that DPC was superior to PC in terms of reducing the incidence of superficial SSI, but with respect to post-op pain and LOS, the two techniques of wound closure were not different.</br>.


Asunto(s)
Apendicitis , Infección de la Herida Quirúrgica , Humanos , Infección de la Herida Quirúrgica/etiología , Apendicitis/cirugía , Estudios Prospectivos , Técnicas de Cierre de Heridas/efectos adversos , Dolor Postoperatorio
15.
J Cereb Blood Flow Metab ; 43(1): 115-128, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36071645

RESUMEN

Cerebral energy deficiency is increasingly recognised as an important feature of multiple sclerosis (MS). Until now, we have lacked non-invasive imaging methods to quantify energy utilisation and mitochondrial function in the human brain. Here, we used novel dual-calibrated functional magnetic resonance imaging (dc-fMRI) to map grey-matter (GM) deoxy-haemoglobin sensitive cerebral blood volume (CBVdHb), cerebral blood flow (CBF), oxygen extraction fraction (OEF), and cerebral metabolic rate of oxygen consumption (CMRO2) in patients with MS (PwMS) and age/sex matched controls. By integrating a flow-diffusion model of oxygen transport, we evaluated the effective oxygen diffusivity of the capillary network (DC) and the partial pressure of oxygen at the mitochondria (PmO2). Significant between-group differences were observed as decreased CBF (p = 0.010), CMRO2 (p < 0.001) and DC (p = 0.002), and increased PmO2 (p = 0.043) in patients compared to controls. No significant differences were observed for CBVdHb (p = 0.389), OEF (p = 0.358), or GM volume (p = 0.302). Regional analysis showed widespread reductions in CMRO2 and DC for PwMS. Our findings may be indicative of reduced oxygen demand or utilisation in the MS brain and mitochondrial dysfunction. Our results suggest changes in brain physiology may precede MRI-detectable GM loss and may contribute to disease progression and neurodegeneration.


Asunto(s)
Esclerosis Múltiple , Humanos , Esclerosis Múltiple/diagnóstico por imagen , Oxígeno , Imagen por Resonancia Magnética , Encéfalo/diagnóstico por imagen
16.
Sci Data ; 10(1): 25, 2023 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-36635305

RESUMEN

The Prostate, Lung, Colorectal and Ovarian (PLCO) Cancer Screening Trial is a prospective cohort study of nearly 155,000 U.S. volunteers aged 55-74 at enrollment in 1993-2001. We developed the PLCO Atlas Project, a large resource for multi-trait genome-wide association studies (GWAS), by genotyping participants with available DNA and genomic consent. Genotyping on high-density arrays and imputation was performed, and GWAS were conducted using a custom semi-automated pipeline. Association summary statistics were generated from a total of 110,562 participants of European, African and Asian ancestry. Application programming interfaces (APIs) and open-source software development kits (SKDs) enable exploring, visualizing and open data access through the PLCO Atlas GWAS Explorer website, promoting Findable, Accessible, Interoperable, and Re-usable (FAIR) principles. Currently the GWAS Explorer hosts association data for 90 traits and >78,000,000 genomic markers, focusing on cancer and cancer-related phenotypes. New traits will be posted as association data becomes available. The PLCO Atlas is a FAIR resource of high-quality genetic and phenotypic data with many potential reuse opportunities for cancer research and genetic epidemiology.


Asunto(s)
Estudio de Asociación del Genoma Completo , Neoplasias Ováricas , Femenino , Humanos , Masculino , Pulmón , Polimorfismo de Nucleótido Simple , Estudios Prospectivos , Próstata
17.
Carcinogenesis ; 33(4): 918-30, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22345290

RESUMEN

Understanding the molecular pathways that contribute to the development of tamoxifen resistance is a critical research priority as acquired tamoxifen resistance is the principal cause of poor prognosis and death of patients with originally good prognosis hormone-responsive breast tumors. In this report, we provide evidence that Med1, an important subunit of mediator coactivator complex, is spontaneously upregulated during acquired tamoxifen-resistance development potentiating agonist activities of tamoxifen. Phosphorylated Med1 and estrogen receptor (ER) are abundant in tamoxifen-resistant breast cancer cells due to persistent activation of extracellular signal-regulated kinases. Mechanistically, phosphorylated Med1 exhibits nuclear accumulation, increased interaction with ER and higher tamoxifen-induced recruitment to ER-responsive promoters, which is abrogated by inhibition of Med1 phosphorylation. Stable knockdown of Med1 in tamoxifen-resistant cells not only reverses tamoxifen resistance in vitro but also in vivo. Finally, higher expression levels of Med1 in the tumor significantly correlated with tamoxifen resistance in ER-positive breast cancer patients on adjuvant tamoxifen monotherapy. In silico analysis of breast cancer, utilizing published profiling studies showed that Med1 is overexpressed in aggressive subsets. These findings provide what we believe is the first evidence for a critical role for Med1 in tamoxifen resistance and identify this coactivator protein as an essential effector of the tamoxifen-induced breast cancer growth.


Asunto(s)
Antineoplásicos Hormonales/farmacología , Resistencia a Antineoplásicos/fisiología , Subunidad 1 del Complejo Mediador/fisiología , Tamoxifeno/farmacología , Western Blotting , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Inmunoprecipitación , Subunidad 1 del Complejo Mediador/metabolismo , Microscopía Fluorescente , Fosforilación , Receptores de Estrógenos/fisiología
19.
Breast Cancer Res ; 14(1): R35, 2012 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-22353783

RESUMEN

INTRODUCTION: Honokiol, a small-molecule polyphenol isolated from magnolia species, is widely known for its therapeutic potential as an antiinflammatory, antithrombosis, and antioxidant agent, and more recently, for its protective function in the pathogenesis of carcinogenesis. In the present study, we sought to examine the effectiveness of honokiol in inhibiting migration and invasion of breast cancer cells and to elucidate the underlying molecular mechanisms. METHODS: Clonogenicity and three-dimensional colony-formation assays were used to examine breast cancer cell growth with honokiol treatment. The effect of honokiol on invasion and migration of breast cancer cells was evaluated by using Matrigel invasion, scratch-migration, spheroid-migration, and electric cell-substrate impedance sensing (ECIS)-based migration assays. Western blot and immunofluorescence analysis were used to examine activation of the liver kinase B1 (LKB1)-AMP-activated protein kinase (AMPK) axis. Isogenic LKB1-knockdown breast cancer cell line pairs were developed. Functional importance of AMPK activation and LKB1 overexpression in the biologic effects of honokiol was examined by using AMPK-null and AMPK-wild type (WT) immortalized mouse embryonic fibroblasts (MEFs) and isogenic LKB1-knockdown cell line pairs. Finally, mouse xenografts, immunohistochemical and Western blot analysis of tumors were used. RESULTS: Analysis of the underlying molecular mechanisms revealed that honokiol treatment increases AMP-activated protein kinase (AMPK) phosphorylation and activity, as evidenced by increased phosphorylation of the downstream target of AMPK, acetyl-coenzyme A carboxylase (ACC) and inhibition of phosphorylation of p70S6kinase (pS6K) and eukaryotic translation initiation factor 4E binding protein 1 (4EBP1). By using AMPK-null and AMPK-WT (MEFs), we found that AMPK is required for honokiol-mediated modulation of pACC-pS6K. Intriguingly, we discovered that honokiol treatment increased the expression and cytoplasmic translocation of tumor-suppressor LKB1 in breast cancer cells. LKB1 knockdown inhibited honokiol-mediated activation of AMPK and, more important, inhibition of migration and invasion of breast cancer cells. Furthermore, honokiol treatment resulted in inhibition of breast tumorigenesis in vivo. Analysis of tumors showed significant increases in the levels of cytoplasmic LKB1 and phospho-AMPK in honokiol-treated tumors. CONCLUSIONS: Taken together, these data provide the first in vitro and in vivo evidence of the integral role of the LKB1-AMPK axis in honokiol-mediated inhibition of the invasion and migration of breast cancer cells. In conclusion, honokiol treatment could potentially be a rational therapeutic strategy for breast carcinoma.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Antineoplásicos/farmacología , Compuestos de Bifenilo/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Lignanos/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Quinasas de la Proteína-Quinasa Activada por el AMP , Proteínas Quinasas Activadas por AMP/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Antineoplásicos/uso terapéutico , Compuestos de Bifenilo/uso terapéutico , Neoplasias de la Mama/patología , Proteínas de Ciclo Celular , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Transformación Celular Neoplásica/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Femenino , Expresión Génica , Humanos , Lignanos/uso terapéutico , Ratones , Ratones Desnudos , Invasividad Neoplásica , Fosfoproteínas/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Quinasas S6 Ribosómicas/metabolismo , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Am J Physiol Gastrointest Liver Physiol ; 302(2): G225-35, 2012 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-22038829

RESUMEN

The aims of this study were designed to determine whether liraglutide, a long-acting glucagon-like peptide, could reverse the adverse effects of a diet high in fat that also contained trans-fat and high-fructose corn syrup (ALIOS diet). Specifically, we examined whether treatment with liraglutide could reduce hepatic insulin resistance and steatosis as well as improve cardiac function. Male C57BL/6J mice were pair fed or fed ad libitum either standard chow or the ALIOS diet. After 8 wk the mice were further subdivided and received daily injections of either liraglutide or saline for 4 wk. Hyperinsulinemic-euglycemic clamp studies were performed after 6 wk, revealing hepatic insulin resistance. Glucose tolerance and insulin resistance tests were performed at 8 and 12 wk prior to and following liraglutide treatment. Liver pathology, cardiac measurements, blood chemistry, and RNA and protein analyses were performed. Clamp studies revealed hepatic insulin resistance after 6 wk of ALIOS diet. Liraglutide reduced visceral adiposity and liver weight (P < 0.001). As expected, liraglutide improved glucose and insulin tolerance. Liraglutide improved hypertension (P < 0.05) and reduced cardiac hypertrophy. Surprisingly, liver from liraglutide-treated mice had significantly higher levels of fatty acid binding protein, acyl-CoA oxidase II, very long-chain acyl-CoA dehydrogenase, and microsomal triglyceride transfer protein. We conclude that liraglutide reduces the harmful effects of an ALIOS diet by improving insulin sensitivity and by reducing lipid accumulation in liver through multiple mechanisms including, transport, and increase ß-oxidation.


Asunto(s)
Cardiomegalia/tratamiento farmacológico , Dieta Alta en Grasa/efectos adversos , Hígado Graso/tratamiento farmacológico , Péptido 1 Similar al Glucagón/análogos & derivados , Adiposidad/efectos de los fármacos , Animales , Cardiomegalia/etiología , Hígado Graso/etiología , Péptido 1 Similar al Glucagón/farmacología , Péptido 1 Similar al Glucagón/uso terapéutico , Prueba de Tolerancia a la Glucosa , Hipertensión/tratamiento farmacológico , Hipertensión/etiología , Resistencia a la Insulina , Liraglutida , Hígado/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA