Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Monaldi Arch Chest Dis ; 84(1-2): 726, 2016 06 22.
Article in English | MEDLINE | ID: mdl-27374040

ABSTRACT

Diabetes mellitus is one of the most important causes of cardiovascular morbidity and mortality; the incidence of chronic complications of diabetes appears to be closely related to the degree of hyperglycaemia. However, results of clinical trials showed that intensive treatment of hyperglycaemia prevents microvascular complications, but has little or no effect on the incidence of cardiovascular events. Different hypoglycaemic drugs show different effects on cardiovascular risk. However, those trials have shown a neutral effect on cardiovascular mortality. This paradoxical result could be explained with the frequent use, in the past, of glucose-lowering agents capable of increasing the risk of hypoglicemia, glycemic variability and weight gain. In conclusion, an adequate glycemic control, in particular in elderly patients, should be achieved, whenever possible, using agents not inducing hypogycemia, glucose fluctuations, and weight gain. In fact, hypoglycaemia and glucose variability should be considered as independent cardiovascular risk factors to a similar extent to hyperglycemia. In this article, the author will review literature supporting the hypothesis that hyperglycemia, hypoglycaemia and glycemic variability are a fatal triad capable of increasing morbidity and mortality in patients with diabetes mellitus.


Subject(s)
Blood Glucose , Cardiovascular Diseases/mortality , Diabetes Mellitus/mortality , Hyperglycemia/mortality , Hypoglycemia/mortality , Hypoglycemic Agents/adverse effects , Aged , Cardiovascular Diseases/etiology , Humans , Hyperglycemia/drug therapy , Hypoglycemia/chemically induced
2.
Am J Physiol Gastrointest Liver Physiol ; 301(2): G210-9, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21252047

ABSTRACT

Leptin modulates the angiogenic properties of hepatic stellate cells (HSC), but the molecular mechanisms involved are poorly understood. We investigated the pathways regulating hypoxia-inducible factor 1α (HIF-1α) and vascular endothelial growth factor (VEGF) in leptin-stimulated myofibroblastic HSC. Exposure to leptin enhanced the phosphorylation of TSC2 on T1462 residues and of p70 S6 kinase and the translational inhibitor 4E-binding protein-1, indicating the ability of leptin to activate the mammalian target of rapamycin (mTOR) pathway. Similar findings were observed when HSC were exposed to PDGF. Both leptin and PDGF increased the expression of HIF-1α and VEGF in HSC. In the presence of rapamycin, a specific mTOR inhibitor, leptin and PDGF were no longer able to activate mTOR, and expression of VEGF was reduced, whereas HIF-1α abundance was not affected. Moreover, knockdown of Raptor, a component of the mTORC1 complex, reduced the ability of leptin to increase VEGF. mTOR was also necessary for leptin- and PDGF-dependent increase in HSC migration. Leptin increased the generation of reactive oxygen species in HSC, which was reduced by NADP(H) oxidase inhibitors. Both N-acetyl cysteine and diphenylene iodonium, a NADP(H) inhibitor, inhibited the expression of HIF-1α and VEGF stimulated by leptin or PDGF. Finally, conditioned media from HSC treated with leptin or PDGF induced tube formation in cultured human umbilical vein endothelial cells. In conclusion, in HSC exposed to leptin or PDGF, increased expression of VEGF requires both activation of mTOR and generation of reactive oxygen species via NADPH-oxidase. Induction of HIF-1α requires NADP(H) oxidase but not mTOR activation.


Subject(s)
Hepatic Stellate Cells/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Leptin/physiology , Liver/blood supply , Signal Transduction/physiology , TOR Serine-Threonine Kinases/physiology , Vascular Endothelial Growth Factor A/metabolism , Cell Line , Cell Movement/physiology , Hep G2 Cells , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Leptin/metabolism , NADPH Oxidases/physiology , Neovascularization, Pathologic , Neovascularization, Physiologic , Phosphorylation , Platelet-Derived Growth Factor/physiology , Reactive Oxygen Species/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/chemistry , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/chemistry , Vascular Endothelial Growth Factor A/physiology
3.
Semin Immunopathol ; 31(3): 345-58, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19533130

ABSTRACT

Fibrosis is a multicellular wound healing process, where myofibroblasts that express extracellular matrix components extensively cross-talk with other cells resident in the liver or recruited from the bloodstream. Macrophages and infiltrating monocytes participate in the development of fibrosis via several mechanisms, including secretion of cytokines and generation of oxidative stress-related products. However, macrophages are also pivotal in the process of fibrosis resolution, where they contribute to matrix degradation. T lymphocytes modulate the fibrogenic process by direct interaction with myofibroblasts and secreting cytokines. In general, Th2 polarized responses promote fibrosis, while Th1 cytokines may be antifibrogenic. NK cells limit the development of fibrosis and favor its resolution, at least in part via killing of fibrogenic cells. The possible role of NKT cells and B cells is emerging in recent studies. Thus, mononuclear cells represent a critical regulatory system during fibrogenesis and may become an appealing target for therapy.


Subject(s)
Cytokines/immunology , Extracellular Matrix/immunology , Liver Cirrhosis/immunology , Toll-Like Receptors/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cytokines/metabolism , Extracellular Matrix/metabolism , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Liver/immunology , Liver/pathology , Liver Cirrhosis/metabolism , Macrophages/immunology , Macrophages/metabolism , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Toll-Like Receptors/metabolism
4.
Hepatology ; 47(2): 668-76, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18098312

ABSTRACT

UNLABELLED: Adiponectin limits the development of liver fibrosis and activates adenosine monophosphate-activated protein kinase (AMPK). AMPK is a sensor of the cellular energy status, but its possible modulation of the fibrogenic properties of hepatic stellate cells (HSCs) has not been established. In this study, we investigated the role of AMPK activation in the biology of activated human HSCs. A time-dependent activation of AMPK was observed in response to a number of stimuli, including globular adiponectin, 5-aminoimidazole-4-carboxamide-1-beta-4-ribofuranoside (AICAR), or metformin. All these compounds significantly inhibited platelet-derived growth factor (PDGF)-stimulated proliferation and migration of human HSCs and reduced the secretion of monocyte chemoattractant protein-1. In addition, AICAR limited the secretion of type I procollagen. Knockdown of AMPK by gene silencing increased the mitogenic effects of PDGF, confirming the negative modulation exerted by this pathway on HSCs. AMPK activation did not reduce PDGF-dependent activation of extracellular signal-regulated kinase (ERK) or Akt at early time points, whereas a marked inhibition was observed 24 hours after addition of PDGF, reflecting a block in cell cycle progression. In contrast, AICAR blocked short-term phosphorylation of ribosomal S6 kinase (p70(S6K)) and 4E binding protein-1 (4EBP1), 2 downstream effectors of the mammalian target of rapamycin (mTOR) pathway, by PDGF. The ability of interleukin-a (IL-1) to activate nuclear factor kappa B (NF-kappaB) was also reduced by AICAR. CONCLUSION: Activation of AMPK negatively modulates the activated phenotype of HSCs.


Subject(s)
Liver/cytology , Liver/physiology , Multienzyme Complexes/metabolism , Protein Serine-Threonine Kinases/metabolism , Stem Cells/cytology , Stem Cells/physiology , AMP-Activated Protein Kinases , Adiponectin/pharmacology , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Chemokine CCL2/metabolism , Chemotaxis/physiology , Enzyme Activation , Humans , Liver/drug effects , Multienzyme Complexes/genetics , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/genetics , Ribonucleotides/pharmacology , Stem Cells/drug effects
5.
J Hepatol ; 46(2): 230-8, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17125873

ABSTRACT

BACKGROUND/AIMS: Administration of carbon tetrachloride determines liver injury, inflammation and oxidative stress, but the molecular mechanisms of damage are only partially understood. In this study, we investigated the development of acute toxic damage in mice lacking monocyte chemoattractant protein-1 (MCP-1), a chemokine which recruits monocytes and activated lymphocytes. METHODS: Mice with targeted deletion of the MCP-1 gene and wild type controls were administered a single intragastric dose of carbon tetrachloride. Serum liver enzymes, histology, expression of different chemokines and cytokines, and intrahepatic levels of oxidative stress-related products were evaluated. RESULTS: Compared to wild type mice, peak aminotransferase levels were significantly lower in MCP-1-deficient animals. This was paralleled by a delayed appearance of necrosis at histology. In addition, MCP-1-deficient mice showed a shift in the pattern of infiltrating inflammatory cells, with a predominance of polymorphonuclear leukocytes. Lack of MCP-1 was also accompanied by reduced intrahepatic expression of cytokines regulating inflammation and tissue repair. The increase in tissue levels of reactive oxygen species and 4-hydroxy-nonenal following administration of the hepatotoxin was also significantly lower in animals lacking MCP-1. CONCLUSIONS: Lack of MCP-1 affords protection from damage and development of oxidative stress in a toxic model of severe acute liver injury.


Subject(s)
Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/immunology , Chemokine CCL2/genetics , Oxidative Stress/genetics , Aldehydes/analysis , Animals , Carbon Tetrachloride/toxicity , Chemical and Drug Induced Liver Injury/prevention & control , Cytokines/genetics , Cytokines/metabolism , Gene Deletion , Liver/drug effects , Liver/pathology , Mice , Mice, Knockout , Neutrophils , Reactive Oxygen Species/analysis , Reactive Oxygen Species/metabolism
6.
Am J Pathol ; 169(6): 2042-53, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17148667

ABSTRACT

Obesity and insulin resistance accelerate the progression of fibrosis during chronic liver disease. Resistin antagonizes insulin action in rodents, but its role in humans is still controversial. The aims of this study were to investigate resistin expression in human liver and to evaluate whether resistin may affect the biology of activated human hepatic stellate cells (HSCs), key modulators of hepatic fibrogenesis. Resistin gene expression was low in normal human liver but was increased in conditions of severe fibrosis. Up-regulation of resistin during chronic liver damage was confirmed by immunohistochemistry. In a group of patients with alcoholic hepatitis, resistin expression correlated with inflammation and fibrosis, suggesting a possible action on HSCs. Exposure of cultured HSCs to recombinant resistin resulted in increased expression of the proinflammatory chemokines monocyte chemoattractant protein-1 and interleukin-8, through activation of nuclear factor (NF)-kappaB. Resistin induced a rapid increase in intracellular calcium concentration, mainly through calcium release from intracellular inositol triphosphate-sensitive pools. The intracellular calcium chelator BAPTA-AM blocked resistin-induced NF-kappaB activation and monocyte chemoattractant protein-1 expression. In conclusion, this study shows a role for resistin as an intrahepatic cytokine exerting proinflammatory actions in HSCs, via a Ca2+/NF-kappaB-dependent pathway and suggests involvement of this adipokine in the pathophysiology of liver fibrosis.


Subject(s)
Inflammation/metabolism , Liver Diseases/metabolism , Liver/metabolism , Resistin/metabolism , Acute Disease , Calcium/metabolism , Case-Control Studies , Cells, Cultured , Chemokines/metabolism , Hepatitis, Alcoholic/metabolism , Humans , Liver/cytology , Liver/immunology , NF-kappa B/metabolism , Resistin/physiology , Signal Transduction , Up-Regulation
7.
Hepatology ; 42(6): 1339-48, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16317688

ABSTRACT

Leptin upregulates collagen expression in hepatic stellate cells (HSCs), but the possible modulation of other actions has not been elucidated. The aim of this study was to investigate the expression and function of leptin receptors (ObR) in human HSCs and the biological actions regulated by leptin. Exposure of HSCs to leptin resulted in upregulation of monocyte chemoattractant protein 1 (MCP-1) expression. Leptin also increased gene expression of the proangiogenic cytokines vascular endothelial growth factor (VEGF) and angiopoietin-1, and VEGF was also upregulated at the protein level. Activated HSCs express ObRb and possibly other ObR isoforms. Exposure to leptin increased the tyrosine kinase activity of ObR immunoprecipitates and resulted in activation of signal transducer and activator of transcription 3. Several signaling pathways were activated by leptin in HSCs, including extracellular-signal-regulated kinase, Akt, and nuclear factor kappaB, the latter being relevant for chemokine expression. Leptin also increased the abundance of hypoxia-inducible factor 1alpha, which regulates angiogenic gene expression, in an extracellular-signal-regulated kinase- and phoshatidylinositol 3-kinase-dependent fashion. In vivo, leptin administration induced higher MCP-1 expression and more severe inflammation in mice after acute liver injury. Conversely, in leptin-deficient mice, the increase in MCP-1 messenger RNA and mononuclear infiltration was less marked than in wild-type littermates. Finally, ObR expression colocalized with VEGF and alpha-smooth muscle actin after induction of fibrosis in rats. In conclusion, ObR activation in HSCs leads to increased expression of proinflammatory and proangiogenic cytokines, indicating a complex role for leptin in the regulation of the liver wound-healing response.


Subject(s)
Chemokine CCL2/genetics , Leptin/pharmacology , Liver Cirrhosis/etiology , Liver/metabolism , Receptors, Cell Surface/physiology , Vascular Endothelial Growth Factor A/genetics , Wound Healing , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Liver/cytology , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Leptin , Signal Transduction , Up-Regulation
8.
Am J Physiol Gastrointest Liver Physiol ; 287(1): G18-26, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15016614

ABSTRACT

Hepatic stellate cells (HSC) coordinate the liver wound-healing response through secretion of several cytokines and chemokines, including CCL2 (formerly known as monocyte chemoattractant protein-1). In this study, we evaluated the role of different proteins of the MAPK family (ERK, p38(MAPK), and JNK) in the regulation of CCL2 expression by HSC, as an index of their proinflammatory activity. Several mediators activated all three MAPK, including TNF, IL-1, and PDGF. To assess the relative role of the different MAPKs, specific pharmacological inhibitors were used; namely, SB203580 (p38(MAPK)), SP600125 (JNK), and PD98059 (MEK/ERK). The efficacy and specificity of the different inhibitors in our cellular system were verified analyzing the enzymatic activity of the different MAPKs using in vitro kinase assays and/or testing the inhibition of phosphorylation of downstream substrates. SB203580 and SP600125 dose-dependently inhibited CCL2 secretion and gene expression induced by IL-1 or TNF. In contrast, inhibition of ERK did not affect the upregulation of CCL2 induced by the two cytokines. Finally, activin A was also found to stimulate CCL2 expression and to activate ERK, JNK, p38, and their downstream targets. Unlike in cells exposed to proinflammatory cytokines, all three MAPKs were required to induce CCL2 secretion in response to activin. We conclude that members of the MAPK family differentially regulate cytokine-induced chemokine expression in human HSC.


Subject(s)
Hepatocytes/metabolism , Mitogen-Activated Protein Kinases/physiology , Activins/pharmacology , Cells, Cultured , Enzyme Inhibitors/pharmacology , Humans , Inhibin-beta Subunits/pharmacology , Interleukin-1/pharmacology , JNK Mitogen-Activated Protein Kinases , Platelet-Derived Growth Factor/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...