Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
2.
Dis Esophagus ; 33(5)2020 May 15.
Article in English | MEDLINE | ID: mdl-31665266

ABSTRACT

Robots are increasingly used in minimally invasive surgery. We evaluated the clinical benefits of robot-assisted minimally invasive esophagectomy (RAMIE) in comparison with the conventional open esophageal surgery. From 2012 to 2016, 371 patients with esophageal squamous cell carcinoma underwent an Ivor Lewis or McKeown procedure at our institution. Of these, 130 patients underwent laparoscopic gastric conduit formation followed by RAMIE, whereas 241 patients underwent conventional esophageal surgery, including laparotomy and open esophagectomy (OE). We compared the short- and long-term clinical outcomes of these patients using the propensity score-based inverse probability of treatment weighting technique (IPTW). Among the early outcomes, the OE group showed a higher incidence of pneumonia (P = 0.035) and a higher requirement for vasopressors (P = 0.001). Regarding the long-term outcomes, all-cause mortality was significantly higher (P = 0.001) and disease-free survival was lower (P = 0.006) in the OE group. Wound-related problems also occurred more frequently in the OE group (P = 0.020) during the long-term follow-up. There was no statistical intergroup difference in the recurrence rates (P = 0.191). The Cox proportional-hazard analysis demonstrated that wound problems (HR 0.16, 95% CI 0.02-0.57; P = 0.017), pneumonia (HR 0.23, 95% CI 0.06-0.68; P = 0.019), and use of vasopressors (HR 0.14, 95% CI 0.08-0.25; P = 0.001) were independent predictors of mortality. RAMIE could be a better surgical option for selected patients with esophageal squamous cell carcinoma.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Head and Neck Neoplasms , Robotic Surgical Procedures , Robotics , Esophageal Neoplasms/surgery , Esophageal Squamous Cell Carcinoma/surgery , Esophagectomy/adverse effects , Humans , Minimally Invasive Surgical Procedures , Neoplasm Recurrence, Local , Postoperative Complications/epidemiology , Postoperative Complications/etiology , Propensity Score , Treatment Outcome
3.
Blood Cancer J ; 6: e387, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26771813

ABSTRACT

KIT exon 17 mutation is a poor prognostic factor in core-binding factor acute myeloid leukemia. However, the mutation detection method used for risk assessment is not assigned. It is necessary to verify the analytical and clinical performance before applying new methods. Herein, we firstly applied a highly sensitive allele-specific, real-time quantitative PCR (AS-qPCR) assay to analyze KIT mutations, which demonstrated excellent sensitivity and specificity. Much higher incidence of KIT mutations (62.2%, 69/111) and prevalence of multiple mutations (43.5%, 30/69) were observed using AS-qPCR, which meant the existence of multiple KIT mutant subclones. The relative KIT mutant level was variable (median, 0.3 per control allele 100 copies, 0.002-532.7) and was divided into two groups: high (⩾10, n=26) and low (<10) mutant level. Interestingly, rather than mutation positivity, mutant level was found to be associated with clinical outcome. High mutant level showed significantly inferior overall survival (P=0.005) and event-free survival (P=0.03), whereas low level did not influence the prognosis. The follow-up data showed that the mutant level were along with fusion transcripts in the majority (n=29), but moved separately in some cases, including the loss of mutations (n=5) and selective proliferation of minor clones (n=2) at relapse. This study highlighted that the KIT mutation should be analyzed using sensitive and quantitative techniques and set a cutoff level for identifying the risk group.


Subject(s)
Core Binding Factors/genetics , Exons , Leukemia, Myeloid, Acute/genetics , Mutation , Proto-Oncogene Proteins c-kit/genetics , Adolescent , Adult , Aged , Alleles , Biopsy , Bone Marrow/pathology , Core Binding Factors/metabolism , Female , Genetic Testing , Humans , Incidence , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/epidemiology , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/therapy , Male , Middle Aged , Real-Time Polymerase Chain Reaction , Sensitivity and Specificity , Sequence Analysis, DNA , Young Adult
4.
J Pathol ; 236(4): 517-30, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25875424

ABSTRACT

Metabolic adaptation is considered an emerging hallmark of cancer, whereby cancer cells exhibit high rates of glucose consumption with consequent lactate production. To ensure rapid efflux of lactate, most cancer cells express high levels of monocarboxylate transporters (MCTs), which therefore may constitute suitable therapeutic targets. The impact of MCT inhibition, along with the clinical impact of altered cellular metabolism during prostate cancer (PCa) initiation and progression, has not been described. Using a large cohort of human prostate tissues of different grades, in silico data, in vitro and ex vivo studies, we demonstrate the metabolic heterogeneity of PCa and its clinical relevance. We show an increased glycolytic phenotype in advanced stages of PCa and its correlation with poor prognosis. Finally, we present evidence supporting MCTs as suitable targets in PCa, affecting not only cancer cell proliferation and survival but also the expression of a number of hypoxia-inducible factor target genes associated with poor prognosis. Herein, we suggest that patients with highly glycolytic tumours have poorer outcome, supporting the notion of targeting glycolytic tumour cells in prostate cancer through the use of MCT inhibitors.


Subject(s)
Glycolysis , Lactic Acid/metabolism , Molecular Targeted Therapy , Monocarboxylic Acid Transporters/metabolism , Prostatic Neoplasms/metabolism , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Proliferation , Disease Progression , Drug Design , Gene Expression Regulation, Neoplastic , Humans , Male , Mice, Transgenic , Monocarboxylic Acid Transporters/antagonists & inhibitors , Monocarboxylic Acid Transporters/genetics , Neoplasm Staging , Phenotype , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA Interference , Time Factors , Transfection , Tumor Burden
5.
Blood Cancer J ; 5: e336, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-26832846

ABSTRACT

Mutation of the fms-like tyrosine kinase 3-internal tandem duplication (FLT3-ITD), which is one of the most frequent genetic alterations, strongly contributes to an increased risk of treatment failure and to poor prognosis. In this study, we established quantitative fragment analysis of FLT3-ITD simultaneously measuring mutant allele burden and length, verified the analytical performance and evaluated the clinical significance in adult acute myeloid leukemia (AML) patients. FLT3-ITD was detected in 73 of 363 adult AML patients (20.1%) and high mutant allelic burden (⩾50%, n=13) and long ITD length (⩾70 base pairs, n=15) were significantly associated with inferior overall survival (OS; P=0.002 and 0.005, respectively) and event-free survival (EFS; P=0.004 and 0.007, respectively). FLT3-ITD poor prognostic group was identified as patients with high allele burden or long ITD length (n=24), which revealed significant adverse clinical outcome for both OS (P<0.001) and EFS (P<0.001). In cytogenetically normal AML, even FLT3-ITD low allele burden and short length was associated with poorer OS (P=0.037) and EFS (P=0.044) than wild type, whose influence was overcome when hematopoietic stem cell transplantation was performed. In minimal residual disease monitoring, FLT3-ITD negativity after consolidation therapy was a valuable predictor of better OS (P<0.001) and EFS (P<0.001). FLT3-ITD poor prognostic group with high mutant allele burden or long ITD length is efficiently identified by quantitative fragment analysis.


Subject(s)
Leukemia, Myeloid, Acute/drug therapy , fms-Like Tyrosine Kinase 3/genetics , Adult , Female , Humans , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/genetics , Male , Middle Aged , Mutation , Polymerase Chain Reaction , Prognosis
6.
Oncotarget ; 5(11): 3785-99, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-25003216

ABSTRACT

Metabolic alterations contribute to prostate cancer development and progression; however, the role of the central metabolic regulator AMP-activated protein kinase (AMPK) remains controversial. The androgen receptor (AR), a key driver of prostate cancer, regulates prostate cancer cell metabolism by driving the expression of a network of metabolic genes and activates AMPK through increasing the expression of one of its upstream kinases. To more clearly define the role of AMPK in prostate cancer, we performed expression profiling following pharmacologic activation of this kinase. We found that genes down-regulated upon AMPK activation were over-expressed in prostate cancer, consistent with a tumour suppressive function of AMPK. Strikingly, we identified the AR as one of the most significantly enriched transcription factors mediating gene expression changes downstream of AMPK signalling in prostate cancer cells. Activation of AMPK inhibited AR transcriptional activity and reduced androgen-dependent expression of known AR target genes. Conversely, knock-down of AMPK increased AR activity. Modulation of AR expression could not explain these effects. Instead, we observed that activation of AMPK reduced nuclear localisation of the AR. We thus propose the presence of a negative feedback loop in prostate cancer cells whereby AR activates AMPK and AMPK feeds back to limit AR-driven transcription.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , Receptors, Androgen/metabolism , Cell Nucleus/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Male , RNA, Small Interfering/genetics , Receptors, Androgen/genetics , Signal Transduction , Transfection , Tumor Cells, Cultured
7.
J Biol Chem ; 289(7): 4244-61, 2014 Feb 14.
Article in English | MEDLINE | ID: mdl-24338480

ABSTRACT

Triglycerides and cholesterol are essential for life in most organisms. Triglycerides serve as the principal energy storage depot and, where vascular systems exist, as a means of energy transport. Cholesterol is essential for the functional integrity of all cellular membrane systems. The endoplasmic reticulum is the site of secretory lipoprotein production and de novo cholesterol synthesis, yet little is known about how these activities are coordinated with each other or with the activity of the COPII machinery, which transports endoplasmic reticulum cargo to the Golgi. The Sar1B component of this machinery is mutated in chylomicron retention disorder, indicating that this Sar1 isoform secures delivery of dietary lipids into the circulation. However, it is not known why some patients with chylomicron retention disorder develop hepatic steatosis, despite impaired intestinal fat malabsorption, and why very severe hypocholesterolemia develops in this condition. Here, we show that Sar1B also promotes hepatic apolipoprotein (apo) B lipoprotein secretion and that this promoting activity is coordinated with the processes regulating apoB expression and the transfer of triglycerides/cholesterol moieties onto this large lipid transport protein. We also show that although Sar1A antagonizes the lipoprotein secretion-promoting activity of Sar1B, both isoforms modulate the expression of genes encoding cholesterol biosynthetic enzymes and the synthesis of cholesterol de novo. These results not only establish that Sar1B promotes the secretion of hepatic lipids but also adds regulation of cholesterol synthesis to Sar1B's repertoire of transport functions.


Subject(s)
Apolipoproteins B/metabolism , Cholesterol/biosynthesis , Endoplasmic Reticulum/metabolism , Lipid Metabolism , Monomeric GTP-Binding Proteins/metabolism , Vesicular Transport Proteins/metabolism , Apolipoproteins B/genetics , COP-Coated Vesicles/genetics , COP-Coated Vesicles/metabolism , Cell Line , Cholesterol/genetics , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/pathology , Golgi Apparatus/genetics , Golgi Apparatus/metabolism , Golgi Apparatus/pathology , Humans , Hypobetalipoproteinemias/genetics , Hypobetalipoproteinemias/metabolism , Hypobetalipoproteinemias/pathology , Lipids/genetics , Liver/metabolism , Liver/pathology , Malabsorption Syndromes/genetics , Malabsorption Syndromes/metabolism , Malabsorption Syndromes/pathology , Monomeric GTP-Binding Proteins/genetics , Vesicular Transport Proteins/genetics
8.
J Lipid Res ; 54(12): 3491-505, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24103848

ABSTRACT

The purpose of this study was to determine the core biological processes perturbed in the subcutaneous adipose tissue of familial combined hyperlipidemia (FCHL) patients. Annotation of FCHL and control microarray datasets revealed a distinctive FCHL transcriptome, characterized by gene expression changes regulating five overlapping systems: the cytoskeleton, cell adhesion and extracellular matrix; vesicular trafficking; lipid homeostasis; and cell cycle and apoptosis. Expression values for the cell-cycle inhibitor CDKN2B were increased, replicating data from an independent FCHL cohort. In 3T3-L1 cells, CDKN2B knockdown induced C/EBPα expression and lipid accumulation. The minor allele at SNP site rs1063192 (C) was predicted to create a perfect seed for the human miRNA-323b-5p. A miR-323b-5p mimic significantly reduced endogenous CDKN2B protein levels and the activity of a CDKN2B 3'UTR luciferase reporter carrying the rs1063192 C allele. Although the allele displayed suggestive evidence of association with reduced CDKN2B mRNA in the MuTHER adipose tissue dataset, family studies suggest the association between increased CDKN2B expression and FCHL-lipid abnormalities is driven by factors external to this gene locus. In conclusion, from a comparative annotation analysis of two separate FCHL adipose tissue transcriptomes and a subsequent focus on CDKN2B, we propose that dysfunctional adipogenesis forms an integral part of FCHL pathogenesis.


Subject(s)
Adipose Tissue/metabolism , Cyclin-Dependent Kinase Inhibitor p15/genetics , Gene Expression Regulation , Hyperlipidemia, Familial Combined/genetics , 3T3-L1 Cells , Adipogenesis/genetics , Adipose Tissue/pathology , Animals , Cell Cycle/genetics , HEK293 Cells , Haplotypes , Humans , Hyperlipidemia, Familial Combined/pathology , Male , Mice , Middle Aged
9.
J Clin Invest ; 117(8): 2325-36, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17671657

ABSTRACT

Hypothalamic AMP-activated protein kinase (AMPK) has been suggested to act as a key sensing mechanism, responding to hormones and nutrients in the regulation of energy homeostasis. However, the precise neuronal populations and cellular mechanisms involved are unclear. The effects of long-term manipulation of hypothalamic AMPK on energy balance are also unknown. To directly address such issues, we generated POMC alpha 2KO and AgRP alpha 2KO mice lacking AMPK alpha2 in proopiomelanocortin- (POMC-) and agouti-related protein-expressing (AgRP-expressing) neurons, key regulators of energy homeostasis. POMC alpha 2KO mice developed obesity due to reduced energy expenditure and dysregulated food intake but remained sensitive to leptin. In contrast, AgRP alpha 2KO mice developed an age-dependent lean phenotype with increased sensitivity to a melanocortin agonist. Electrophysiological studies in AMPK alpha2-deficient POMC or AgRP neurons revealed normal leptin or insulin action but absent responses to alterations in extracellular glucose levels, showing that glucose-sensing signaling mechanisms in these neurons are distinct from those pathways utilized by leptin or insulin. Taken together with the divergent phenotypes of POMC alpha 2KO and AgRP alpha 2KO mice, our findings suggest that while AMPK plays a key role in hypothalamic function, it does not act as a general sensor and integrator of energy homeostasis in the mediobasal hypothalamus.


Subject(s)
Energy Metabolism/physiology , Homeostasis/physiology , Hypothalamus/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Multienzyme Complexes/metabolism , Neurons/metabolism , Pro-Opiomelanocortin/metabolism , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases , Agouti-Related Protein , Animals , Eating/physiology , Glucose/metabolism , Insulin/metabolism , Intercellular Signaling Peptides and Proteins/deficiency , Leptin/metabolism , Mice , Mice, Knockout , Multienzyme Complexes/deficiency , Pro-Opiomelanocortin/deficiency , Protein Serine-Threonine Kinases/deficiency , Signal Transduction/physiology
10.
Am J Transplant ; 7(2): 480-3, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17173654

ABSTRACT

Patient safety in transplantation depends on accurate testing, transcription and transmission of the ABO types of the donor and recipient. Similar to 'near-miss' transfusion labeling errors, three cases of mislabeled ABO types on deceased donor kidney containers were recognized through a pretransplant verification process. Six steps to confirming the organ and ABO identification were developed to ensure safety of the patient and prevent liability for the transplant team and facility. In each case, rapid recognition and documentation of the error source, on site confirmation of the ABO type of the accompanying blood specimen, and full disclosure to the patient and family permitted safe transplantation and avoided the need to pursue a more conservative course that would have required discarding the organs. We advocate following these measures in determining whether to persevere with transplantation of a mislabeled organ.


Subject(s)
ABO Blood-Group System/immunology , Kidney Transplantation/immunology , Medical Errors/prevention & control , Product Labeling/standards , Aged , Blood Group Incompatibility/diagnosis , Blood Group Incompatibility/immunology , Humans , Male , Safety Management/methods
11.
J Biol Chem ; 279(16): 15719-22, 2004 Apr 16.
Article in English | MEDLINE | ID: mdl-14970221

ABSTRACT

The mammalian target of rapamycin (mTOR) is a key regulator of protein translation. Signaling via mTOR is increased by growth factors but decreased during nutrient deprivation. Previous studies have identified Ser2448 as a nutrient-regulated phosphorylation site located in the mTOR catalytic domain, insulin stimulates Ser2448 phosphorylation via protein kinase B (PKB), while Ser2448 phosphorylation is attenuated with amino acid starvation. Here we have identified Thr2446 as a novel nutrient-regulated phosphorylation site on mTOR. Thr2446 becomes phosphorylated when CHO-IR cells are nutrient-deprived, but phosphorylation is reduced by insulin stimulation. Nutrient deprivation activates AMP-activated protein kinase (AMPK). To test whether this could be involved in regulating phoshorylation of mTOR, we treated cultured murine myotubes with 5'-aminoimidazole-4-carboxamide ribonucleoside (AICAR) or dinitrophenol (DNP). Both treatments activated AMPK and also caused a concomitant increase in phosphorylation of Thr2446 and a parallel decrease in insulin's ability to phosphorylate p70 S6 kinase. In vitro kinase assays using peptides based on the sequence in amino acids 2440-2551 of mTOR found that PKB and AMPK are capable of phosphorylating sites in this region. However, phosphorylation by PKB is restricted when Thr2446 is mutated to an acidic residue mimicking phosphorylation. Conversely, AMP-kinase-induced phosphorylation is reduced when Ser2448 is phosphorylated. These data suggest differential phosphorylation Thr2446 and Ser2448 could act as a switch mechanism to integrate signals from nutrient status and growth factors to control the regulation of protein translation.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Protein Kinases/metabolism , Signal Transduction , Sirolimus/metabolism , AMP-Activated Protein Kinases , Aminoimidazole Carboxamide/pharmacology , Animals , CHO Cells , Cricetinae , Dinitrophenols/pharmacology , Enzyme Activation/drug effects , Humans , Insulin/metabolism , Mice , Multienzyme Complexes/metabolism , Phosphorylation , Protein Biosynthesis , Protein Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Ribonucleotides/pharmacology , TOR Serine-Threonine Kinases , Threonine/metabolism
12.
J Biol Chem ; 279(13): 12260-8, 2004 Mar 26.
Article in English | MEDLINE | ID: mdl-14711829

ABSTRACT

Neuregulin-1, a growth factor that potentiates myogenesis induces glucose transport through translocation of glucose transporters, in an additive manner to insulin, in muscle cells. In this study, we examined the signaling pathway required for a recombinant active neuregulin-1 isoform (rhHeregulin-beta(1), 177-244, HRG) to stimulate glucose uptake in L6E9 myotubes. The stimulatory effect of HRG required binding to ErbB3 in L6E9 myotubes. PI3K activity is required for HRG action in both muscle cells and tissue. In L6E9 myotubes, HRG stimulated PKBalpha, PKBgamma, and PKCzeta activities. TPCK, an inhibitor of PDK1, abolished both HRG- and insulin-induced glucose transport. To assess whether PKB was necessary for the effects of HRG on glucose uptake, cells were infected with adenoviruses encoding dominant negative mutants of PKBalpha. Dominant negative PKB reduced PKB activity and insulin-stimulated glucose transport but not HRG-induced glucose transport. In contrast, transduction of L6E9 myotubes with adenoviruses encoding a dominant negative kinase-inactive PKCzeta abolished both HRG- and insulin-stimulated glucose uptake. In soleus muscle, HRG induced PKCzeta, but not PKB phosphorylation. HRG also stimulated the activity of p70S6K, p38MAPK, and p42/p44MAPK and inhibition of p42/p44MAPK partially repressed HRG action on glucose uptake. HRG did not affect AMPKalpha(1) or AMPKalpha(2) activities. In all, HRG stimulated glucose transport in muscle cells by activation of a pathway that requires PI3K, PDK1, and PKCzeta, but not PKB, and that shows cross-talk with the MAPK pathway. The PI3K, PDK1, and PKCzeta pathway can be considered as an alternative mechanism, independent of insulin, to induce glucose uptake.


Subject(s)
Glucose/metabolism , Muscle Cells/metabolism , Neuregulin-1/metabolism , Signal Transduction , 3-Phosphoinositide-Dependent Protein Kinases , Adenoviridae/genetics , Adenylate Kinase/metabolism , Animals , Biological Transport , Cell Line , Enzyme Inhibitors/pharmacology , Genes, Dominant , Glucose/pharmacokinetics , Immunoblotting , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Muscle, Skeletal/metabolism , Muscles/metabolism , Oncogene Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Precipitin Tests , Protein Binding , Protein Isoforms , Protein Kinase C/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Synthesis Inhibitors/pharmacology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Recombinant Proteins/chemistry , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Time Factors , Tosylphenylalanyl Chloromethyl Ketone/pharmacology , p38 Mitogen-Activated Protein Kinases
13.
Curr Biol ; 13(22): 2004-8, 2003 Nov 11.
Article in English | MEDLINE | ID: mdl-14614828

ABSTRACT

Inactivating mutations in the protein kinase LKB1 lead to a dominantly inherited cancer in humans termed Peutz-Jeghers syndrome. The role of LKB1 is unclear, and only one target for LKB1 has been identified in vivo [3]. AMP-activated protein kinase (AMPK) is the downstream component of a protein kinase cascade that plays a pivotal role in energy homeostasis. AMPK may have a role in protecting the body from metabolic diseases including type 2 diabetes, obesity, and cardiac hypertrophy. We previously reported the identification of three protein kinases (Elm1, Pak1, and Tos3 [9]) that lie upstream of Snf1, the yeast homologue of AMPK. LKB1 shares sequence similarity with Elm1, Pak1, and Tos3, and we demonstrated that LKB1 phosphorylates AMPK on the activation loop threonine (Thr172) within the catalytic subunit and activates AMPK in vitro [9]. Here, we have investigated whether LKB1 corresponds to the major AMPKK activity present in cell extracts. AMPKK purified from rat liver corresponds to LKB1, and blocking LKB1 activity in cells abolishes AMPK activation in response to different stimuli. These results identify a link between two protein kinases, previously thought to lie in unrelated, distinct pathways, that are associated with human diseases.


Subject(s)
Multienzyme Complexes/metabolism , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases , Animals , Blotting, Western , Chromatography, Ion Exchange , Enzyme Activation , Liver/metabolism , Precipitin Tests , Rats
14.
J Physiol ; 546(Pt 2): 327-35, 2003 Jan 15.
Article in English | MEDLINE | ID: mdl-12527721

ABSTRACT

We determined whether mitogen-activated protein kinase (MAPK) and 5'-AMP-activated protein kinase (AMPK) signalling cascades are activated in response to intense exercise in skeletal muscle from six highly trained cyclists (peak O(2) uptake (.V(O2,peak)) 5.14 +/- 0.1 l min(-1)) and four control subjects (Vdot;(O(2))(,peak) 3.8 +/- 0.1 l min(-1)) matched for age and body mass. Trained subjects completed eight 5 min bouts of cycling at approximately 85% of .V(O2,peak) with 60 s recovery between work bouts. Control subjects performed four 5 min work bouts commencing at the same relative, but a lower absolute intensity, with a comparable rest interval. Vastus lateralis muscle biopsies were taken at rest and immediately after exercise. Extracellular regulated kinase (ERK1/2), p38 MAPK, histone H3, AMPK and acetyl CoA-carboxylase (ACC) phosphorylation was determined by immunoblot analysis using phosphospecific antibodies. Activity of mitogen and stress-activated kinase 1 (MSK1; a substrate of ERK1/2 and p38 MAPK) and alpha(1) and alpha(2) subunits of AMPK were determined by immune complex assay. ERK1/2 and p38 MAPK phosphorylation and MSK1 activity increased (P < 0.05) after exercise 2.6-, 2.1- and 2.0-fold, respectively, in control subjects and 1.5-, 1.6- and 1.4-fold, respectively, in trained subjects. Phosphorylation of histone H3, a substrate of MSK1, increased (P < 0.05) approximately 1.8-fold in both control and trained subject. AMPKalpha(2) activity increased (P < 0.05) after exercise 4.2- and 2.3-fold in control and trained subjects, respectively, whereas AMPKalpha(1) activity was not altered. Exercise increased ACC phosphorylation (P < 0.05) 1.9- and 2.8-fold in control and trained subjects. In conclusion, intense cycling exercise in subjects with a prolonged history of endurance training increases MAPK signalling to the downstream targets MSK1 and histone H3 and isoform-specific AMPK signalling to ACC. Importantly, exercise-induced signalling responses were greater in untrained men, even at the same relative exercise intensity, suggesting muscle from previously well-trained individuals requires a greater stimulus to activate signal transduction via these pathways.


Subject(s)
Bicycling/physiology , Mitogen-Activated Protein Kinases/metabolism , Multienzyme Complexes/metabolism , Muscle, Skeletal/metabolism , Physical Endurance , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , AMP-Activated Protein Kinases , Acetyl-CoA Carboxylase/metabolism , Adult , Histones/metabolism , Humans , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Phosphorylation , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , p38 Mitogen-Activated Protein Kinases
15.
Diabetes ; 52(1): 9-15, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12502487

ABSTRACT

We report here use of human myoblasts in culture to study the relationships between cellular glycogen concentrations and the activities of glycogen synthase (GS) and AMP-activated protein kinase (AMPK). Incubation of cells for 2 h in the absence of glucose led to a 25% decrease in glycogen content and a significant decrease in the fractional activity of GS. This was accompanied by stimulation of both the alpha1 and alpha2 isoforms of AMPK, without significant alterations in the ratios of adenine nucleotides. When glucose was added to glycogen-depleted cells, a rapid and substantial increase in GS activity was accompanied by inactivation of AMPK back to basal values. Inclusion of the glycogen phosphorylase inhibitor, CP-91149, prevented the loss of glycogen during glucose deprivation but not the activation of AMPK. However, in the absence of prior glycogen breakdown, glucose treatment failed to activate GS above control values, indicating the crucial role of glycogen content. Activation of AMPK by either 5-aminoimidazole-4-carboxamide 1-beta-D-ribofuranoside (AICAR) or hydrogen peroxide was also associated with a decrease in the activity ratio of GS. AICAR treatment had no effect on total cellular glycogen content but led to a modest increase in glucose uptake. These data support a role for AMPK in both stimulating glucose uptake and inhibiting GS in intact cells, thus promoting glucose flux through glycolysis.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Glucose/pharmacology , Glycogen Synthase/metabolism , Glycogen/pharmacology , AMP-Activated Protein Kinases , Amides/pharmacology , Aminoimidazole Carboxamide/pharmacology , Cells, Cultured , Enzyme Activation/drug effects , Glucose/deficiency , Glycogen Phosphorylase/antagonists & inhibitors , Glycogen Synthase/antagonists & inhibitors , Humans , Hydrogen Peroxide/pharmacology , Indoles/pharmacology , Isoenzymes/metabolism , Multienzyme Complexes/physiology , Myoblasts/enzymology , Protein Serine-Threonine Kinases/physiology , Ribonucleotides/pharmacology
16.
FEBS Lett ; 531(2): 189-92, 2002 Nov 06.
Article in English | MEDLINE | ID: mdl-12417310

ABSTRACT

The AMP-activated protein kinase (AMPK) is the central component of a protein kinase cascade that plays a major role in energy sensing. AMPK is activated pharmacologically by 5-amino-4-imidazolecarboxamide (AICA) riboside monophosphate (ZMP), which mimics the effects of AMP on the AMPK cascade. Here we show that uptake of AICA riboside into cells, mediated by the adenosine transport system, is blocked by a number of protein kinase inhibitors. Under these conditions, ZMP does not accumulate to sufficient levels to stimulate AMPK. Our results demonstrate that careful interpretation is required when using AICA riboside in conjunction with protein kinase inhibitors to investigate the physiological role of AMPK.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Enzyme Inhibitors/pharmacology , Multienzyme Complexes/metabolism , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Ribonucleosides/pharmacology , AMP-Activated Protein Kinases , Adenosine/metabolism , Animals , Biological Transport/drug effects , Cells, Cultured , Enzyme Activation , Kinetics , Muscle, Skeletal/drug effects , Muscle, Skeletal/enzymology , Ribonucleotides/biosynthesis
17.
Diabetes ; 51(9): 2703-8, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12196462

ABSTRACT

Glucose transport can be activated in skeletal muscle in response to insulin via activation of phosphoinositide (PI) 3-kinase and in response to contractions or hypoxia, presumably via activation of 5' AMP-activated protein kinase (AMPK). We determined the effects of insulin and muscle contraction/hypoxia on PI 3-kinase, AMPK, and glucose transport activity in epitrochlearis skeletal muscle from insulin-resistant Zucker (fa/ fa) rats. Insulin-stimulated glucose transport in isolated skeletal muscle was reduced 47% in obese versus lean rats, with a parallel 42% reduction in tyrosine-associated PI 3-kinase activity. Contraction and hypoxia elicited normal responses for glucose transport in skeletal muscle from insulin-resistant obese rats. Isoform-specific AMPK activity was measured in skeletal muscle in response to insulin, contraction, or hypoxia. Contraction increased AMPKalpha1 activity 2.3-fold in lean rats, whereas no effect was noted in obese rats. Hypoxia increased AMPKalpha1 activity to a similar extent (more than sixfold) in lean and obese rats. Regardless of genotype, contraction, and hypoxia, each increased AMPKalpha2 activity more than fivefold, whereas insulin did not alter either AMPKalpha1 or -alpha2 activity in skeletal muscle. In conclusion, obesity-related insulin resistance is associated with an isoform-specific impairment in AMPKalpha1 in response to contraction. However, this impairment does not appear to affect contraction-stimulated glucose transport. Activation of AMPKalpha2 in response to muscle contraction/ exercise is associated with a parallel and normal increase in glucose transport in insulin-resistant skeletal muscle.


Subject(s)
Adenosine Monophosphate/physiology , Muscle Contraction/physiology , Muscle, Skeletal/enzymology , Obesity/enzymology , Obesity/physiopathology , Protein Kinases/metabolism , Animals , Glucose/pharmacokinetics , Hypoxia/metabolism , Isoenzymes/metabolism , Male , Phosphatidylinositol 3-Kinases/metabolism , Rats , Rats, Zucker
18.
Tissue Antigens ; 59(5): 430-2, 2002 May.
Article in English | MEDLINE | ID: mdl-12144630

ABSTRACT

We have identified a new HLA-B*15 allele and a new HLA-DRB1*12 allele, named B*1568 and DRB1*1208, respectively. The alleles were identified using a combination of sequence specific primers, reverse line sequence specific oligonucleotide probing and sequence-based typing. Both alleles were identified in a single individual of Korean origin. HLA-B*1568 appears to be an HLA-B*4801/B*1507 hybrid combining the exon 2 sequence of B*4801 and the exon 3 and 4 sequences of B*1507. Exon 2 of DRB1*1208 was most similar to DRB1*1201 or 1206, with a single mismatch at nucleotide position 165 (A to C). At the protein level, this substitution results in a phenylalanine substitution at position 26 that creates an identical amino acid sequence to DRB3*0202 between amino acid positions 17 and 36.


Subject(s)
HLA-B Antigens/genetics , HLA-DR Antigens/genetics , Amino Acid Sequence , Exons , HLA-B15 Antigen , HLA-DRB1 Chains , Humans , Korea , Molecular Sequence Data
19.
J Cell Sci ; 115(Pt 11): 2433-42, 2002 Jun 01.
Article in English | MEDLINE | ID: mdl-12006627

ABSTRACT

In the rat liver epithelial cell line Clone 9, the V(max) for glucose uptake is acutely increased by inhibition of oxidative phosphorylation and by osmotic stress. By using a membrane-impermeant photoaffinity labelling reagent together with an isoform-specific antibody, we have, for the first time, provided direct evidence for the involvement of the GLUT1 glucose transporter isoform in this response. Transport stimulation was found to be associated with enhanced accessibility of GLUT1 to its substrate and with photolabelling of formerly 'cryptic' exofacial substrate binding sites in GLUT1 molecules. The total amount of cell surface GLUT1 remained constant. The precise mechanism for this binding site 'unmasking' is unclear but appears to involve AMP-activated protein kinase: in the current study, osmotic and metabolic stresses were found to result in activation of the alpha 1 isoform of AMP-activated protein kinase, and transport stimulation could be mimicked both by 5-aminoimidazole-4-carboxamide ribonucleoside and by infection of cells with a recombinant adenovirus encoding constitutively active AMP-activated protein kinase. The effect of 5-aminoimidazole-4-carboxamide ribonucleoside, as for metabolic stress, was on the V(max) rather than on the K(m) for transport and did not affect the cell-surface concentration of GLUT1. The relevant downstream target(s) of AMP-activated protein kinase have not yet been identified, but stimulation of transport by inhibition of oxidative phosphorylation or by 5-aminoimidazole-4-carboxamide ribonucleoside was not prevented by either inhibitors of conventional and novel protein kinase C isoforms or inhibitors of nitric oxide synthase. These enzymes, which have been implicated in stress-regulated pathways in other cell types, are therefore unlikely to play a role in transport regulation by stress in Clone 9 cells.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Glucose/metabolism , Hepatocytes/enzymology , Monosaccharide Transport Proteins/metabolism , Multienzyme Complexes/metabolism , Osmotic Pressure , Protein Serine-Threonine Kinases/metabolism , Stress, Physiological/metabolism , AMP-Activated Protein Kinases , Aminoimidazole Carboxamide/pharmacology , Animals , Cells, Cultured , Enzyme Inhibitors/pharmacology , Glucose Transporter Type 1 , Hepatocytes/cytology , Hypoglycemic Agents/pharmacology , Monosaccharide Transport Proteins/drug effects , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Oxidative Phosphorylation/drug effects , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Rats , Ribonucleotides/pharmacology
20.
J Biol Chem ; 277(28): 25226-32, 2002 Jul 12.
Article in English | MEDLINE | ID: mdl-11994296

ABSTRACT

AMP-activated protein kinase (AMPK) is activated within the cell in response to multiple stresses that increase the intracellular AMP:ATP ratio. Here we show that incubation of muscle cells with the thiazolidinedione, rosiglitazone, leads to a dramatic increase in this ratio with the concomitant activation of AMPK. This finding raises the possibility that a number of the beneficial effects of the thiazolidinediones could be mediated via activation of AMPK. Furthermore, we show that in addition to the classical activation pathway, AMPK can also be stimulated without changing the levels of adenine nucleotides. In muscle cells, both hyperosmotic stress and the anti-diabetic agent, metformin, activate AMPK in the absence of any increase in the AMP:ATP ratio. However, although activation is no longer dependent on this ratio, it still involves increased phosphorylation of threonine 172 within the catalytic (alpha) subunit. AMPK stimulation in response to hyperosmotic stress does not appear to involve phosphatidylinositol 3-phosphate kinase, protein kinase C, mitogen-activated protein (MAP) kinase kinase, or p38 MAP kinase alpha or beta. Our results demonstrate that AMPK can be activated by at least two distinct signaling mechanisms and suggest that it may play a wider role in the cellular stress response than was previously understood.


Subject(s)
Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Multienzyme Complexes/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Thiazoles/pharmacology , Thiazolidinediones , AMP-Activated Protein Kinases , Animals , Catalysis , Enzyme Activation , Mice , Mice, Transgenic , Phosphorylation , Rosiglitazone
SELECTION OF CITATIONS
SEARCH DETAIL
...