Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
JCI Insight ; 7(18)2022 09 22.
Article in English | MEDLINE | ID: mdl-35862222

ABSTRACT

Although published studies have demonstrated that IFN-ε has a crucial role in regulating protective immunity in the mouse female reproductive tract, expression and regulation of IFN-ε in the human female reproductive tract (hFRT) have not been characterized to our knowledge. We obtained hFRT samples from a well-characterized cohort of women to enable us to comprehensively assess ex vivo IFN-ε expression in the hFRT at various stages of the menstrual cycle. We found that among the various types of IFNs, IFN-ε was uniquely, selectively, and constitutively expressed in the hFRT epithelium. It had distinct expression patterns in the surface and glandular epithelia of the upper hFRT compared with basal layers of the stratified squamous epithelia of the lower hFRT. There was cyclical variation of IFN-ε expression in the endometrial epithelium of the upper hFRT and not in the distal FRT, consistent with selective endometrial expression of the progesterone receptor and regulation of the IFNE promoter by progesterone. Because we showed IFN-ε stimulated important protective IFN-regulated genes in FRT epithelium, this characterization is a key element in understanding the mechanisms of hormonal control of mucosal immunity.


Subject(s)
Endometrium , Immunity, Innate , Interferons , Animals , Endometrium/immunology , Epithelium/immunology , Female , Gene Expression Regulation , Humans , Immunity, Innate/genetics , Interferons/genetics , Interferons/metabolism , Mice , Progesterone/metabolism , Promoter Regions, Genetic , Receptors, Progesterone/metabolism
2.
J Clin Diagn Res ; 9(5): PR01-3, 2015 May.
Article in English | MEDLINE | ID: mdl-26155519

ABSTRACT

Right lower quadrant pain is one of the major reasons of children reference at the emergency department. The most common surgical cause, which needs appropriate management, is acute appendicitis. The purpose of this study is to reveal uncommon surgical causes found during surgery in children who were misdiagnosed as acute appendicitis in our department during the last 10 y. Data of patients who have undergone appendicectomy during a ten year period (since Feb 2004 until Mar 2014) were collected retrospectively. Eight hundred twenty children have undergone appendicectomy in our department. In six children another uncommon cause of the symptoms was revealed during surgery. In one patient the cause was a duplication cyst of the terminal ileum, in two patients an omental torsion, in one patient a meckel diverticulum torsion, in one patient a splenic rupture and in one patient a retroperitoneal tumor. All of the patients were successfully managed during the first operation. The possibility of other uncommon causes of right quadrant abdominal pain should always be kept in mind, especially when there is a negative appendicitis. However, the transaction of further paraclinical examinations - ultrasonography or computed tomography- preoperatively is under discussion. Nevertheless a thoroughly taken case history is undoubtedly always necessary.

3.
Urologia ; 82(1): 71-3, 2015.
Article in English | MEDLINE | ID: mdl-24832471

ABSTRACT

The aim of this study is to present a novel use of Dextranomer/hyaluronic acid copolymer (Deflux) for the treatment of a complicated iatrogenic chronic urethral injury. A 12-year-old boy with a neuropathic bladder presented a groove in the urethral mucosa due to chronic clean intermittent catheterizations (CICs) and suffered a posterior urethral injury during a failed catheterization for a routine examination. The defect on the urethral wall was repaired using Deflux, a technique that is not described in the literature. After the intervention the patient is continuing CICs without further complications.


Subject(s)
Biocompatible Materials/administration & dosage , Dextrans/administration & dosage , Hyaluronic Acid/administration & dosage , Urethra/injuries , Urinary Bladder, Neurogenic/etiology , Urinary Bladder, Neurogenic/therapy , Urinary Catheterization/adverse effects , Urinary Catheters/adverse effects , Child , Cystoscopy/methods , Humans , Male , Meningomyelocele/complications , Treatment Outcome
4.
JOP ; 15(3): 250-3, 2014 May 27.
Article in English | MEDLINE | ID: mdl-24865536

ABSTRACT

CONTEXT: Optimal management of penetrating pancreaticoduodenal injuries and better outcomes are associated with simple, fast damage control surgery and shorter operative time. The performance of pyloric exclusion and tube duodenostomy has markedly decreased. However, there is still a trend toward their performance in cases of delay duodenal repair or severe pancreaticoduodenal injury. CASE REPORT: The present report describes a case of a hemodynamically stable patient with a single penetrating gunshot trauma causing an AAST-OIS grade III pancreatic head injury and grade IV injury of the second portion of the duodenum. The patient was treated in our Level IV rural trauma center and submitted to primary closure of the posterolateral duodenal wall (the laceration of the contralateral inner medial duodenal wall could not be repaired), external duodenal and pancreatic drainage, and duodenal decompression by tube pancreatico-duodenostomy (insertion of a 18 Fr Foley catheter through the laceration of the pancreatic head toward the duodenal lumen), tube cholangiostomy, and pyloric exclusion accompanied with a feeding jejunostomy. CONCLUSIONS: Tube pancreatico-duodenostomy, which is described for the first time in the literature, turned out to be effective and can be considered as an option in pancreaticoduodenal trauma when the inner medial duodenal wall cannot be repaired.


Subject(s)
Duodenostomy/methods , Hemostatic Techniques , Pancreaticoduodenectomy/methods , Trauma Severity Indices , Wounds, Gunshot/surgery , Adult , Duodenum/injuries , Duodenum/surgery , Hemoperitoneum/surgery , Humans , Male , Pancreas/injuries , Pancreas/surgery , Suture Techniques , Treatment Outcome
5.
Updates Surg ; 66(1): 51-7, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24254381

ABSTRACT

Radiation-induced afferent loop obstruction is a rare complication following pancreaticoduodenectomy and adjuvant radiotherapy. As in the setting of Roux-en-Y reconstruction endoscopic approaches are limited, surgery of this complication becomes inevitable. This study provides a new classification/management system of the radiation-induced obstruction of the afferent loop based on the extent and location of radiation injury, and describes the Pancreaticojejuno-jejunostomy, a novel technique to avoid revision of the pancreatic anastomosis during reconstruction of the afferent loop. Data were analyzed from nine patients who developed radiation-induced afferent loop obstruction after pancreaticoduodenectomy with single Roux limb reconstruction. One patient had type I obstruction and treated with by-pass surgery, seven patients had type II obstruction and treated with reconstruction including revision of the hepaticojejunostomy and Pancreaticojejuno-jejunostomy, and one patient had type III obstruction and treated with reconstruction including revision of the hepaticojejunostomy and the pancreatic anastomosis. Reconstruction along with Pancreaticojejuno-jejunostomy performed in six patients with type II radiation-induced afferent loop obstruction; reconstruction was not feasible for one patient. The median operative time was 149 min. No intraoperative complication was observed. By performing Pancreaticojejuno-jejunostomy we managed efficiently to convert a pancreatic anastomosis to an enteric anastomosis as one case of Grade B pancreatic fistula and no case of Pancreaticojejuno-jejunostomy stricture were observed, regarding short- and long-term results, respectively. The above technique may have a useful application in the surgical management of the radiation-induced afferent loop obstruction when endoscopy fails and by-pass surgery is inappropriate.


Subject(s)
Afferent Loop Syndrome/surgery , Pancreatic Neoplasms/surgery , Pancreaticoduodenectomy/adverse effects , Pancreaticojejunostomy/methods , Plastic Surgery Procedures/methods , Afferent Loop Syndrome/etiology , Aged , Aged, 80 and over , Female , Humans , Jejunostomy , Male , Middle Aged , Radiation Injuries/surgery
6.
J Mol Cell Cardiol ; 56: 106-15, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23277190

ABSTRACT

Cardiotrophin-1 (CT-1) is a heart-targeting cytokine that is increased in the metabolic syndrome due to overexpression in the adipocytes. The effects of CT-1 on cardiomyocyte substrate metabolism remain unknown. We therefore determined the effects of CT-1 on basal and stimulated glucose transport in cardiomyocytes exposed to a low dose (1nM) or a high dose (10nM). Dose-response curves for insulin showed that 1nM CT-1 reduced insulin responsiveness, while 10nM CT-1 increased insulin responsiveness. In either condition insulin sensitivity was unaffected. Similarly 1nM CT-1 reduced the stimulation of glucose transport in response to metabolic stress, induced by the mitochondrial poison oligomycin, while 10nM CT-1 increased this response. Reduction of stimulated glucose transport by 1nM CT-1 was associated with overexpression of SOCS-3, a protein known to hinder proximal insulin signaling, and increased phosphorylation of STAT5. In cardiomyocytes exposed to 1nM CT-1 there was also reduced phosphorylation of Akt and AS160 in response to insulin, and of AMPK in response to oligomycin. Insulin-stimulated glucose transport and signaling were restored by inhibition of STAT5 activity. On the other hand in cardiomyocytes exposed to 10nM CT-1 there was increased phosphorylation of the AS160 and Akt in response to insulin. Most importantly, basal and oligomycin-stimulated phosphorylation of AMPK was markedly increased in cardiomyocytes exposed to 10nM CT-1. The enhancement of basal and stimulated-glucose transport was abolished in cardiomyocytes treated with the calmodulin-dependent kinase II (CaMKII) inhibitor KN93, and so was AMPK phosphorylation. This suggests that activation of CaMKII mediates activation of AMPK by a high dose of CT-1 independently of metabolic stress. Our results point to a role for CT-1 in the regulation of myocardial glucose metabolism and implicate entirely separate mechanisms in the inhibitory or stimulatory effects of CT-1 on glucose transport at low or high concentrations respectively.


Subject(s)
Cytokines/physiology , Glucose/metabolism , Myocytes, Cardiac/metabolism , Animals , Biological Transport , Cell Hypoxia , Cells, Cultured , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 4/metabolism , Insulin/physiology , Male , Oligomycins/pharmacology , Phosphorylation , Protein Processing, Post-Translational , Pyruvate Dehydrogenase (Lipoamide)/metabolism , Rats , Rats, Sprague-Dawley , STAT5 Transcription Factor/antagonists & inhibitors , STAT5 Transcription Factor/metabolism , Signal Transduction , Stress, Physiological
7.
Am J Physiol Heart Circ Physiol ; 302(9): H1795-805, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22408021

ABSTRACT

Normal myocardium adapts to increase of nutritional fatty acid supply by upregulation of regulatory proteins of the fatty acid oxidation pathway. Because advanced heart failure is associated with reduction of regulatory proteins of fatty acid oxidation, we hypothesized that failing myocardium may not be able to adapt to increased fatty acid intake and therefore undergo lipid accumulation, potentially aggravating myocardial dysfunction. We determined the effect of high-fat diet in transgenic mice with overexpression of angiotensinogen in the myocardium (TG1306/R1). TG1306/R1 mice develop ANG II-mediated left ventricular hypertrophy, and at one year of age approximately half of the mice present heart failure associated with reduced expression of regulatory proteins of fatty acid oxidation and reduced palmitate oxidation during ex vivo working heart perfusion. Hypertrophied hearts from TG1306/R1 mice without heart failure adapted to high-fat feeding, similarly to hearts from wild-type mice, with upregulation of regulatory proteins of fatty acid oxidation and enhancement of palmitate oxidation. There was no myocardial lipid accumulation or contractile dysfunction. In contrast, hearts from TG1306/R1 mice presenting heart failure were unable to respond to high-fat feeding by upregulation of fatty acid oxidation proteins and enhancement of palmitate oxidation. This resulted in accumulation of triglycerides and ceramide in the myocardium, and aggravation of contractile dysfunction. In conclusion, hearts with ANG II-induced contractile failure have lost the ability to enhance fatty acid oxidation in response to increased fatty acid supply. The ensuing accumulation of lipid compounds may play a role in the observed aggravation of contractile dysfunction.


Subject(s)
Angiotensin II/metabolism , Diet, High-Fat , Dietary Fats/pharmacology , Heart Failure/metabolism , Lipid Metabolism/drug effects , Myocardium/metabolism , Ventricular Remodeling/physiology , Angiotensinogen/genetics , Angiotensinogen/metabolism , Animals , Fatty Acids/metabolism , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/physiopathology , Male , Mice , Mice, Transgenic , Models, Animal , Myocardial Contraction/drug effects , Oxidation-Reduction , Palmitates/metabolism , Triglycerides/metabolism
8.
Am J Physiol Endocrinol Metab ; 302(7): E872-84, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22297301

ABSTRACT

Stimulation of glucose transport in response to insulin or metabolic stress is an important determinant of cardiac myocyte function and survival, particularly during ischemia-reperfusion episodes. The impact of dyslipidemia and its consequence PPAR activation on stimulated glucose transport in cardiac myocytes remains unknown. Isolated adult rat cardiac myocytes were chronically exposed to free fatty acids (FFA) or PPAR agonists. Insulin- (ISGT) and oligomycin-stimulated glucose transport (OSGT) and related cell signaling were analyzed. Exposure of cardiac myocytes to FFA reduced both ISGT and OSGT. Exposure to either PPARα or PPARδ agonists, but not to a PPARγ agonist, reduced ISGT but not OSGT and increased fatty acid oxidation (FAO). The reduction in ISGT was associated with impaired insulin signaling and, in the case of PPAR stimulation, overexpression of SOCS-3, a protein known to hinder proximal insulin signaling. In contrast, the reduction of OSGT could not be explained by a reduced activity of the cellular energy-sensing system, as assessed from the maintained phosphorylation state of AMPK. Inhibition of FAO at the level of mitochondrial acylcarnitine uptake restored OSGT but not ISGT. Seemingly paradoxically, further stimulation of FAO with PPARα or PPARδ agonists also restored OSGT but not ISGT. Together, these results suggest that inhibition of OSGT occurs downstream of energy gauging and is caused by some intermediate(s) of fatty acid oxidation, which does not appear to be acylcarnitines. The results indicate that the mechanisms underlying FFA-mediated inhibition of ISGT and OSGT differ remarkably.


Subject(s)
Biological Transport, Active/drug effects , Fatty Acids, Nonesterified/pharmacology , Glucose/metabolism , Myocytes, Cardiac/metabolism , PPAR alpha/agonists , PPAR delta/agonists , Animals , Antimetabolites/metabolism , Blotting, Western , Cells, Cultured , Deoxyglucose/metabolism , Glucose Transport Proteins, Facilitative/biosynthesis , Glucose Transport Proteins, Facilitative/genetics , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Male , Microscopy, Confocal , Myocytes, Cardiac/drug effects , Oligomycins/pharmacology , Oxidation-Reduction , Palmitates/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Signal Transduction/drug effects , Uncoupling Agents/pharmacology
9.
Am J Physiol Endocrinol Metab ; 301(5): E836-43, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21771966

ABSTRACT

We previously demonstrated that microtubule disruption impairs stimulation of glucose uptake in cardiomyocytes and that 9-cis retinoic acid (9cRA) treatment preserved both microtubule integrity and stimulated glucose transport. Herein we investigated whether 1) activation of the extracellular signal-regulated kinases (ERK1/2) is responsible for microtubule destabilization and 2) ERK1/2 inactivation may explain the positive effects of 9cRA on glucose uptake and microtubule stabilization. Adult rat cardiomyocytes in primary culture showed increased basal ERK1/2 phosphorylation. Cardiomyocytes exposed to inhibitors of the ERK1/2 kinase mitogen/extracellular signal-regulated kinase (MEK) 1/2 had preserved microtubular scaffold, including microtubule-organizing centers (MTOC), together with increased insulin and metabolic stress-stimulated glucose transport as well as signaling, thus replicating the effects of 9cRA treatment. Although 9cRA treatment did not significantly reduce global ERK1/2 activation, it markedly reduced perinuclear-activated ERK1/2 at the location of MTOC. 9cRA also triggered relocation of the ERK1/2 phosphatase mitogen-activated protein kinase phosphatase-3 from the cytosol to the nucleus. These results indicate that, in cardiomyocytes, microtubule destabilization, leading to impaired stimulation of glucose transport, is mediated by ERK1/2 activation, impacting on the MTOC. 9cRA acid restores stimulated glucose transport indirectly through compartmentalized inactivation of ERK1/2.


Subject(s)
Glucose/metabolism , MAP Kinase Signaling System/physiology , Microtubules/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myocytes, Cardiac/metabolism , Alitretinoin , Animals , Biological Transport/drug effects , Biological Transport/physiology , Cells, Cultured , Enzyme Activation/drug effects , Enzyme Activation/physiology , Insulin/pharmacology , MAP Kinase Signaling System/drug effects , Male , Microtubules/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Primary Cell Culture , Protein Multimerization/drug effects , Protein Stability/drug effects , Rats , Rats, Sprague-Dawley , Stress, Physiological/physiology , Tretinoin/pharmacology
10.
Reprod Biol Endocrinol ; 7: 122, 2009 Oct 29.
Article in English | MEDLINE | ID: mdl-19874624

ABSTRACT

BACKGROUND: The human endometrium is unique in its capacity to remodel constantly throughout adult reproductive life. Although the processes of tissue damage and breakdown in the endometrium have been well studied, little is known of how endometrial regeneration is achieved after menstruation. Nodal, a member of the transforming growth factor-beta superfamily, regulates the processes of pattern formation and differentiation that occur during early embryo development. METHODS: In this study, the expression of Nodal, Cripto (co-receptor) and Lefty A (antagonist) was examined by RT-PCR and immunohistochemistry across the menstrual cycle and in endometrial carcinomas. RESULTS: Nodal and Cripto were found to be expressed at high levels in both stromal and epithelial cells during the proliferative phase of the menstrual cycle. Although immunoreactivity for both proteins in surface and glandular epithelium was maintained at relatively steady-state levels across the cycle, their expression was significantly decreased within the stromal compartment by the mid-secretory phase. Lefty expression, as has previously been reported, was primarily restricted to glandular epithelium and surrounding stroma during the late secretory and menstrual phases. In line with recent studies that have shown that Nodal pathway activity is upregulated in many human cancers, we found that Nodal and Cripto immunoreactivity increased dramatically in the transition from histologic Grade 1 to histologic Grades 2 and 3 endometrial carcinomas. Strikingly, Lefty expression was low or absent in all cancer tissues. CONCLUSION: The expression of Nodal in normal and malignant endometrial cells that lack Lefty strongly supports an important role for this embryonic morphogen in the tissue remodelling events that occur across the menstrual cycle and in tumourogenesis.


Subject(s)
Carcinoma/genetics , Endometrial Neoplasms/genetics , Endometrium/metabolism , Menstrual Cycle/genetics , Nodal Protein/genetics , Adult , Body Fluids/metabolism , Carcinoma/metabolism , Endometrial Neoplasms/metabolism , Endometrium/physiology , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , Female , GPI-Linked Proteins , Gene Expression Regulation, Neoplastic , Humans , Intercellular Signaling Peptides and Proteins , Left-Right Determination Factors/genetics , Left-Right Determination Factors/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Menstrual Cycle/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nodal Protein/metabolism , Nodal Protein/physiology , Signal Transduction/genetics , Uterus/metabolism
11.
World J Surg Oncol ; 7: 47, 2009 May 19.
Article in English | MEDLINE | ID: mdl-19454018

ABSTRACT

BACKGROUND: Mucinous cystic neoplasms arise in the ovary and various extra-ovarian sites. While their pathogenesis remains conjectural, their similarities suggest a common pathway of development. There have been rare reports involving the mesentery as a primary tumour site. CASE PRESENTATION: A cystic mass of uncertain origin was demonstrated radiologically in a 22 year old female with chronic abdominal pain. At laparotomy, the mass was fixed within the colonic mesentery. Histology demonstrated a benign mucinous cystadenoma. METHODS AND RESULTS: We review the literature on mucinous cystic neoplasms of the mesentery and report on the pathogenesis, biologic behavior, diagnosis and treatment of similar extra-ovarian tumors. We propose an updated classification of mesenteric cysts and cystic tumors. CONCLUSION: Mucinous cystic neoplasms of the mesentery present almost exclusively in women and must be considered in the differential diagnosis of mesenteric tumors. Only full histological examination of a mucinous cystic neoplasm can exclude a borderline or malignant component. An updated classification of mesenteric cysts and cystic tumors is proposed.


Subject(s)
Cystadenoma, Mucinous/pathology , Mesenteric Cyst/pathology , Adult , Cystadenoma, Mucinous/diagnosis , Cystadenoma, Mucinous/etiology , Cystadenoma, Mucinous/therapy , Female , Humans , Mesenteric Cyst/diagnosis , Mesenteric Cyst/etiology , Mesenteric Cyst/therapy
12.
Cardiovasc Res ; 82(2): 341-50, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19131364

ABSTRACT

AIMS: Advanced heart failure is often associated with reduced myocardial fatty acid oxidation capacity. We have previously observed that failing hearts of mice with overexpression of angiotensinogen in the myocardium exhibit marked reduction of key regulatory proteins of fatty acid oxidation. In the present study, we determined whether exposure of adult rat cardiac (ARC) myocytes to angiotensin II (Ang II) influences expression of fatty acid translocase, muscle-type carnitine palmitoyl transferase-I, and medium-chain acyl-CoA dehydrogenase. METHODS AND RESULTS: Ang II reduced mRNA expression of the three regulatory proteins in ARC myocytes during the entire 14-days culture period. However, protein expression and palmitate oxidation rate remained unaltered for 7 days, but subsequently markedly decreased. The decrease of protein expression and of fatty acid oxidation coincided with the onset of increased protein expression of tumour necrosis factor-alpha (TNF-alpha). The effect of Ang II was completely abolished by either blocking TNF-alpha formation through inhibition of reactive oxygen species-mediated activation of nuclear factor-kappaB or by neutralizing TNF-alpha with a specific antibody. Activation of peroxisome proliferator-activated receptor-alpha (PPARalpha) and PPARbeta/delta counteracted Ang II-mediated reduction of the fatty acid oxidation pathway. CONCLUSION: Prolonged exposure of cardiac myocytes to Ang II elicits downregulation of the fatty acid oxidation pathway mediated by enhanced synthesis of TNF-alpha.


Subject(s)
Angiotensin II/pharmacology , Fatty Acids/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism , Acyl-CoA Dehydrogenase/metabolism , Animals , Carnitine O-Palmitoyltransferase/metabolism , Cells, Cultured , Male , Myocytes, Cardiac/cytology , NF-kappa B/metabolism , Natriuretic Peptide, Brain/metabolism , Oxidation-Reduction , PPAR alpha/metabolism , PPAR delta/metabolism , PPAR gamma/metabolism , PPAR-beta/metabolism , Rats , Rats, Inbred Strains , Reactive Oxygen Species/metabolism
13.
Endocrinology ; 149(3): 1064-74, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18063688

ABSTRACT

Insulin resistance is the failure of insulin to stimulate the transport of glucose into its target cells. A highly regulatable supply of glucose is important for cardiomyocytes to cope with situations of metabolic stress. We recently observed that isolated adult rat cardiomyocytes become insulin resistant in vitro. Insulin resistance is combated at the whole body level with agonists of the nuclear receptor complex peroxisome proliferator-activated receptor gamma (PPARgamma)/retinoid X receptor (RXR). We investigated the effects of PPARgamma/RXR agonists on the insulin-stimulated glucose transport and on insulin signaling in insulin-resistant adult rat cardiomyocytes. Treatment of cardiomyocytes with ciglitazone, a PPARgamma agonist, or 9-cis retinoic acid (RA), a RXR agonist, increased insulin- and metabolic stress-stimulated glucose transport, whereas agonists of PPARalpha or PPARbeta/delta had no effect. Stimulation of glucose transport in response to insulin requires the phosphorylation of the signaling intermediate Akt on the residues Thr308 and Ser473 and, downstream of Akt, AS160 on several Thr and Ser residues. Phosphorylation of Akt and AS160 in response to insulin was lower in insulin-resistant cardiomyocytes. However, treatment with 9-cis RA markedly increased phosphorylation of both proteins. Treatment with 9-cis RA also led to better preservation of microtubules in cultured cardiomyocytes. Disruption of microtubules in insulin-responsive cardiomyocytes abolished insulin-stimulated glucose transport and reduced phosphorylation of AS160 but not Akt. Metabolic stress-stimulated glucose transport also involved AS160 phosphorylation in a microtubule-dependent manner. Thus, the stimulation of glucose uptake in response to insulin or metabolic stress is dependent in cardiomyocytes on the presence of intact microtubules.


Subject(s)
Insulin Resistance/physiology , Insulin/physiology , Myocytes, Cardiac/physiology , PPAR gamma/agonists , Retinoid X Receptors/agonists , Signal Transduction/physiology , AMP-Activated Protein Kinase Kinases , Alitretinoin , Animals , Cells, Cultured , Cytoskeleton/drug effects , Cytoskeleton/physiology , Cytoskeleton/ultrastructure , Glucose/metabolism , Glucose Transporter Type 4/metabolism , Male , Microtubules/drug effects , Microtubules/physiology , Microtubules/ultrastructure , Myocytes, Cardiac/drug effects , PPAR gamma/physiology , Phenoxyacetates/pharmacology , Protein Kinases/pharmacology , Protein Kinases/physiology , Pyrimidines/pharmacology , Rats , Rats, Sprague-Dawley , Retinoid X Receptors/physiology , Signal Transduction/drug effects , Thiazolidinediones/pharmacology , Tretinoin/pharmacology
14.
Pflugers Arch ; 455(3): 443-54, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17643263

ABSTRACT

Inactivation of peroxisome proliferator-activated receptor (PPARs) isoforms alpha, beta/delta, and gamma mediate distinct facets of hypertrophic transformation of adult cardiac myocytes. PPARs are ligand-activated transcription factors that modulate the transcriptional regulation of fatty acid metabolism and the hypertrophic response in neonatal cardiac myocytes. The purpose of this study was to determine the role of PPAR isoforms in the morphologic and metabolic phenotype transformation of adult cardiac myocytes in culture, which, in medium containing 20% fetal calf serum, undergo hypertrophy-like cell growth associated with downregulation of regulatory proteins of fatty acid metabolism. Expression and DNA-binding activity of PPARalpha, PPARbeta/delta, and PPARgamma rapidly decreased after cell isolation and remained persistently reduced during the 14-day culture period. Cells progressively increased in size and developed both re-expression of atrial natriuretic factor and downregulation of regulatory proteins of fatty acid metabolism. Supplementation of the medium with fatty acid (oleate 0.25 mM/palmitate 0.25 mM) prevented inactivation of PPARs and downregulation of metabolic genes. Furthermore, cell size and markers of hypertrophy were markedly reduced. Selective activation of either PPARalpha or PPARbeta/delta completely restored expression of regulatory genes of fatty acid metabolism but did not influence cardiac myocyte size and markers of hypertrophy. Conversely, activation of PPARgamma prevented cardiomyocyte hypertrophy but had no effect on fatty acid metabolism. The results indicate that PPAR activity markedly influences hypertrophic transformation of adult rat cardiac myocytes. Inactivation of PPARalpha and PPARbeta/delta accounts for downregulation of the fatty acid oxidation pathway, whereas inactivation of PPARgamma enables development of hypertrophy.


Subject(s)
Cardiomegaly/etiology , Myocytes, Cardiac/physiology , PPAR alpha/physiology , PPAR delta/physiology , PPAR gamma/physiology , PPAR-beta/physiology , Acyl-CoA Dehydrogenase/biosynthesis , Animals , Atrial Natriuretic Factor/biosynthesis , CD36 Antigens/biosynthesis , Cardiomegaly/physiopathology , Carnitine O-Palmitoyltransferase/biosynthesis , Cells, Cultured , Male , Oleic Acid/pharmacology , PPAR alpha/agonists , PPAR delta/agonists , PPAR gamma/agonists , PPAR-beta/agonists , Palmitates/pharmacology , Phenoxyacetates/pharmacology , Pyrimidines/pharmacology , RNA, Messenger/metabolism , Rats , Thiazolidinediones/pharmacology
15.
J Mol Cell Cardiol ; 42(6): 1016-25, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17512536

ABSTRACT

Myocardial remodeling late after infarction is associated with increased incidence of fatal arrhythmias. Heterogeneous prolongation of the action potential in the surviving myocardium is one of the predominant causes. Sarcolemmal ATP-dependent potassium (K(ATP)) channels are important metabolic sensors regulating electrical activity of cardiomyocytes and are capable of considerably shortening the action potential. We determined whether ATP-dependent potassium channels generate or, on the contrary prevent the heterogeneity in action potential prolongation. Cardiomyocytes were obtained from the infarct border zone, the septum and the right ventricle of rat hearts 20 weeks after coronary occlusion when rats developed signs of heart failure. Expression of the conductance subunit Kir6.1, but not Kir6.2, and of all SUR regulatory subunits was increased up to 3-fold in cardiomyocytes from the infarct border zone. Concomitantly, there was a prominent response of the K(ATP) current to diazoxide that was not detectable in control cardiomyocytes. The action potential was prolonged in cardiomyocytes from the infarct border zone (74 ms) relative to sham (41 ms). However, activation of the K(ATP) channels by diazoxide reduced action potential duration to 42 ms. In myocytes of the septum and right ventricle, expression of channel subunits, duration of action potential, and sensitivity to diazoxide were only slightly increased relative to shams. In conclusion, the myocardium remodeled after infarction displays alterations of K(ATP) expression and function with spatial heterogeneity matching that of the action potential prolongation. Drugs selectively activating diazoxide-sensitive sarcolemmal K(ATP) channels should be considered in the prevention of arrhythmias in post-infarction heart failure.


Subject(s)
Adenosine Triphosphate/metabolism , Myocardial Infarction/physiopathology , Potassium Channels, Inwardly Rectifying/metabolism , Ventricular Remodeling , Action Potentials/drug effects , Animals , Diazoxide/pharmacology , Electric Capacitance , Gene Expression , Heart Septum/cytology , Heart Ventricles/cytology , KATP Channels , Male , Myocardial Infarction/complications , Myocardial Infarction/etiology , Myocytes, Cardiac/metabolism , Patch-Clamp Techniques , Potassium Channels, Inwardly Rectifying/drug effects , Potassium Channels, Inwardly Rectifying/genetics , Rats , Rats, Inbred Strains , Time Factors , Vasodilator Agents/pharmacology
16.
J Mol Cell Cardiol ; 41(3): 459-66, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16859699

ABSTRACT

Heart failure is associated with downregulation of the fatty acid oxidation pathway in the ventricular myocardium. Since angiotensin II plays a critical role in myocardial phenotypic changes associated with heart failure, we investigated the effect of chronic angiotensin II stimulation on the fatty acid oxidation pathway in transgenic (TG) mice with targeted overexpression of angiotensinogen in the myocardium (TG1306/1R mice). TG1306/R1 mice progressively developed left ventricular hypertrophy. After 12 months, approximately half of the mice exhibited signs of heart failure including increased lung weight index [>+2 SD of age-matched wild-type (WT) mice] and 5-fold increase of myocardial brain natriuretic peptide expression. Myocardial mRNA and protein expression of peroxisome proliferator-activated receptor alpha (PPARalpha) progressively decreased in both WT and TG1306/R1 mice during the 12 months observation period, but much more pronounced in TG1306/R1 mice. Concomitantly, mRNA expression of enzymes of fatty acid oxidation (medium-chain acyl CoA dehydrogenase, MCAD; carnitine palmitoyl transferase I, CPT-I) was reduced in TG1306/R1 compared with age-matched WT mice. However, protein expression of MCAD and CPT-I was decreased concomitantly only in TG mice with criteria of heart failure. Correspondingly, myocardial oxidation of palmitate, measured during ex vivo working heart perfusion, was reduced by 25% in TG1306/R1 mice with heart failure. These results demonstrate that angiotensin II-induced cardiac hypertrophy is associated with reduction of PPARalpha and of mRNA expression of enzymes of fatty acid metabolism relative to age-matched WT mice. However, both protein expression of fatty acid oxidation enzymes and the rate of fatty acid oxidation remain unchanged unless heart failure occurs, suggesting the involvement of posttranscriptional mechanisms in the metabolic changes associated with heart failure.


Subject(s)
Angiotensinogen/biosynthesis , Down-Regulation , Fatty Acids/metabolism , Myocardium/metabolism , Animals , Biological Transport , Heart Failure/metabolism , Hypertrophy , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , PPAR alpha/metabolism , Phenotype , RNA, Messenger/metabolism
17.
Basic Res Cardiol ; 101(1): 27-35, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16273324

ABSTRACT

Sufficient expression of the insulin-sensitive glucose transporter GLUT4 may be crucial for the survival of cardiac myocytes in situations of stress. Expression of GLUT4 in cardiac myocytes correlates with cell differentiation and is reduced in the hypertrophied and failing myocardium. Adult rat cardiomyocytes (ARC) in primary culture undergo dedifferentiation and reduction of GLUT4 expression. Depending on the culture condition partial redifferentiation and/or hypertrophy follows. All-trans (at) and 9-cis retinoic acids (RA) are morphogenetic agents important for cell differentiation. Both atRA and 9-cisRA restored GLUT4 expression in dedifferentiated ARC, while only 9-cisRA could increase GLUT4 expression in hypertrophic ARC. The effects of RA were associated with improved differentiation of the cardiac myocytes, as assessed from the expression of atrial natriuretic factor and the morphology of the contractile apparatus. In neonatal rat cardiomyocytes, 9-cisRA, but not atRA, stimulated transcription from the glut4 promoter. In conclusion, treatment with RA can restore the down-regulated expression of GLUT4 in cardiomyocytes in association with a partial improvement of the differentiated phenotype.


Subject(s)
Cardiomyopathy, Hypertrophic/metabolism , Glucose Transporter Type 4/metabolism , Myocytes, Cardiac/metabolism , Tretinoin/physiology , Animals , Cell Differentiation , Cells, Cultured , Male , Myocytes, Cardiac/cytology , Rats , Rats, Sprague-Dawley , Transcription, Genetic , Ventricular Remodeling
18.
Cardiovasc Res ; 64(1): 94-104, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15364617

ABSTRACT

OBJECTIVE: In vivo differentiation of cardiac myocytes is associated with downregulation of the glucose transporter isoform GLUT1 and upregulation of the isoform GLUT4. Adult rat cardiomyocytes in primary culture undergo spontaneous dedifferentiation, followed by spreading and partial redifferentiation, which can be influenced by growth factors. We used this model to study the signaling mechanisms modifying the expression of GLUT4 in cardiac myocytes. RESULTS: Adult rat cardiomyocytes in primary culture exhibited spontaneous upregulation of GLUT1 and downregulation of GLUT4, suggesting resumption of a fetal program of GLUT gene expression. Treatment with IGF-1 and, to a minor extent, FGF-2 resulted in restored expression of GLUT4 protein and mRNA. Activation of p38 MAPK mediated the increased expression of GLUT4 in response to IGF-1. Transient transfection experiments in neonatal cardiac myocytes confirmed that p38 MAPK could activate the glut4 promoter. Electrophoretic mobility shift assay in adult rat cardiomyocytes and transient transfection experiments in neonatal cardiac myocytes indicated that MEF2 was the main transcription factor transducing the effect of p38 MAPK activation on the glut4 promoter. CONCLUSION: Spontaneous dedifferentiation of adult rat cardiomyocytes in vitro is associated with downregulation of GLUT4, which can be reversed by treatment with IGF-1. The effect of IGF-1 is mediated by the p38 MAPK/MEF2 axis, which is a strong inducer of GLUT4 expression.


Subject(s)
MAP Kinase Signaling System/physiology , Monosaccharide Transport Proteins/metabolism , Muscle Proteins/metabolism , Myocytes, Cardiac/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Animals, Newborn , Cells, Cultured , Electrophoretic Mobility Shift Assay , Enzyme Activation , Fibroblast Growth Factor 2/analysis , Fibroblast Growth Factor 2/metabolism , Fluorescent Antibody Technique , Genetic Engineering , Glucose Transporter Type 1 , Glucose Transporter Type 4 , Immunoblotting/methods , Insulin-Like Growth Factor I/analysis , Insulin-Like Growth Factor I/metabolism , Luciferases/genetics , Male , Mitogen-Activated Protein Kinases/metabolism , Monosaccharide Transport Proteins/analysis , Monosaccharide Transport Proteins/genetics , Muscle Proteins/analysis , Muscle Proteins/genetics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
19.
FASEB J ; 18(7): 872-4, 2004 May.
Article in English | MEDLINE | ID: mdl-15117888

ABSTRACT

Myocardium undergoing remodeling in vivo exhibits insulin resistance that has been attributed to a shift from the insulin-sensitive glucose transporter GLUT4 to the fetal, less insulin-sensitive, isoform GLUT1. To elucidate the role of altered GLUT4 expression in myocardial insulin resistance, glucose uptake and the expression of the glucose transporter isoforms GLUT4 and GLUT1 were measured in adult rat cardiomyocytes (ARC). ARC in culture spontaneously undergo dedifferentiation, hypertrophy-like spreading, and return to a fetal-like gene expression pattern. Insulin stimulation of 2-deoxy-D-glucose uptake was completely abolished on day 2 and 3 of culture and recovered thereafter. Although GLUT4 protein level was reduced, the time-course of unresponsiveness to insulin did not correlate with altered expression of GLUT1 and GLUT4. However, translocation of GLUT4 to the sarcolemma in response to insulin was completely abolished during transient insulin resistance. Insulin-mediated phosphorylation of Akt was not reduced, indicating that activation of phosphatidylinositol 3-kinase (PI3K) was preserved. On the other hand, total and phosphorylated Cbl was reduced during insulin resistance, suggesting that activation of Cbl/CAP is essential for insulin-mediated GLUT4 translocation, in addition to activation of PI3K. Pharmacological inhibition of contraction in insulin-sensitive ARC reduced insulin sensitivity and lowered phosphorylated Cbl. The results suggest that transient insulin resistance in ARC is related to impairment of GLUT4 translocation. A defect in the PI3K-independent insulin signaling pathway involving Cbl seems to contribute to reduced insulin responsiveness and may be related to contractile arrest.


Subject(s)
Diacetyl/analogs & derivatives , Insulin Resistance , Monosaccharide Transport Proteins/physiology , Muscle Proteins , Myocytes, Cardiac/drug effects , Phosphatidylinositol 3-Kinases/physiology , Protein Transport/physiology , Proto-Oncogene Proteins/physiology , Ubiquitin-Protein Ligases/physiology , Animals , Atrial Natriuretic Factor/biosynthesis , Atrial Natriuretic Factor/genetics , Biological Transport, Active/drug effects , Calcium Channel Blockers/pharmacology , Cell Differentiation , Cell Size , Cells, Cultured/cytology , Cells, Cultured/drug effects , Deoxyglucose/metabolism , Diacetyl/pharmacology , Enzyme Activation , Gene Expression Regulation , Glucose Transporter Type 1 , Glucose Transporter Type 4 , Insulin/pharmacology , Insulin Resistance/physiology , Monosaccharide Transport Proteins/biosynthesis , Monosaccharide Transport Proteins/genetics , Myocardial Contraction/drug effects , Myocytes, Cardiac/cytology , Phosphorylation , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-cbl , RNA, Messenger/biosynthesis , Rats , Sarcolemma/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Verapamil/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...