Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Minerva Cardiol Angiol ; 69(4): 449-457, 2021 08.
Article in English | MEDLINE | ID: mdl-33258564

ABSTRACT

BACKGROUND: Fractional flow reserve (FFR) is the "gold standard" for assessing the physiological significance of coronary disease. In the last decade, several alternative adenosine-free indexes have been proposed in order to facilitate the dissemination of the functional evaluation of coronary stenosis. Our aim was to investigate whether radiographic contrast plus intracoronary nitroglycerin (cFFR-NTG) can predict functional assessment of coronary stenosis offering superior diagnostic agreement with FFR compared to non-hyperemic indexes and contrast mediated FFR (cFFR). METHODS: Three hundred twenty-nine lesions evaluated with pressure wire in 266 patients were prospectively included in this multicenter study. RESULTS: The ROC curves for cFFR-NTG using an FFR≤0.80 showed a higher accuracy in predicting FFR (AUC=0.97) than resting Pd/Pa (AUC=0.90, P<0.01) and cFFR (AUC=0.93.5, P<0.01). A significant (P<0.01) strong correlation was found between FFR and the four analyzed indexes: Pd/Pa (r=0.78); iFR/RFR (r=0.73); cFFR(r=0.89) and cFFR-NTG (r=0.93). cFFR-NTG showed the closest agreement at Bland-Altman analysis. The cFFR-NTG cut off value >0.84 showed the highest negative predictive value (88%), specificity (91%), sensitivity (94%) and accuracy (92%) of the studied indexes. CONCLUSIONS: Submaximal hyperemic adenosine-free indexes are an efficient alternative to adenosine for the physiological assessment of epicardial coronary disease. The most accurate index in predicting the functional significance of coronary stenosis using FFR as reference was cFFR-NTG.


Subject(s)
Coronary Artery Disease , Coronary Stenosis , Fractional Flow Reserve, Myocardial , Coronary Stenosis/diagnosis , Glycerol , Humans , Prospective Studies
2.
Neuroscience ; 404: 445-458, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30708048

ABSTRACT

Neurotrophic factors (NTFs) are a relevant group of secreted proteins that modulate growth, differentiation, repair, and survival of neurons, playing a role in the maintenance of the synaptic unions, dendrites, and axons and also being crucial for peripheral nervous system development and regulating plasticity in the adult central nervous system. On the other hand, insulin-like growth factor 1 (IGF-1) has been ascertained multiple beneficial actions in the brain: neuro-development, -protection, -genesis and plasticity. To further investigate the possible mechanisms underlying IGF-1 deficiency in the establishment of neurological disease, microarray and reverse transcription polymerase chain reaction gene expression analyses coupled with in silico processing were performed in an experimental model of partial IGF-1 deficiency. Results show that the mere IGF-1 deficiency seems to be responsible for an altered expression of genes coding for neurotrophic factors (particularly ciliary neurotrophic factor and mesencephalic astrocyte-derived neurotrophic factor), their receptors and signaling pathways (specially RET). The presented findings support that IGF-1 deficiency might be involved in the establishment and progression of neurodegenerative disorders.


Subject(s)
Brain/metabolism , Insulin-Like Growth Factor I/deficiency , Nerve Growth Factors/metabolism , Receptors, Nerve Growth Factor/metabolism , Animals , Base Sequence , Insulin-Like Growth Factor I/genetics , Male , Mice , Mice, Transgenic , Nerve Growth Factors/genetics , Receptors, Nerve Growth Factor/genetics
3.
PLoS One ; 12(8): e0181760, 2017.
Article in English | MEDLINE | ID: mdl-28806738

ABSTRACT

Circulating levels of IGF-1 may decrease under several circumstances like ageing, metabolic syndrome, and advanced cirrhosis. This reduction is associated with insulin resistance, dyslipidemia, progression to type 2 diabetes, and increased risk for cardiovascular diseases. However, underlying mechanisms between IGF-1 deficiency and cardiovascular disease remain elusive. The specific aim of the present work was to study whether the partial IGF-1 deficiency influences heart and/or coronary circulation, comparing vasoactive factors before and after of ischemia-reperfusion (I/R). In addition, histology of the heart was performed together with cardiac gene expression for proteins involved in structure and function (extracellular matrix, contractile proteins, active peptides); carried out using microarrays, followed by RT-qPCR confirmation of the three experimental groups. IGF-1 partial deficiency is associated to a reduction in contractility and angiotensin II sensitivity, interstitial fibrosis as well as altered expression pattern of genes involved in extracellular matrix proteins, calcium dynamics, and cardiac structure and function. Although this work is descriptive, it provides a clear insight of the impact that partial IGF-1 deficiency on the heart and establishes this experimental model as suitable for studying cardiac disease mechanisms and exploring therapeutic options for patients under IGF-1 deficiency conditions.


Subject(s)
Angiotensin II/pharmacology , Gene Expression Regulation/drug effects , Insulin-Like Growth Factor I/deficiency , Myocardial Contraction/drug effects , Animals , Body Weight/drug effects , Bradykinin/pharmacology , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Hemodynamics/drug effects , Insulin-Like Growth Factor I/metabolism , Mice, Transgenic , Myocardial Contraction/genetics , Myocardium/metabolism , Myocardium/pathology , Organ Size/drug effects , Perfusion , Real-Time Polymerase Chain Reaction , Vasoconstriction/drug effects , Vasodilation/drug effects
4.
J Transl Med ; 15(1): 96, 2017 05 04.
Article in English | MEDLINE | ID: mdl-28472963

ABSTRACT

BACKGROUND: Cell necrosis, oxidative damage, and fibrogenesis are involved in cirrhosis development, a condition in which insulin-like growth factor 1 (IGF-1) levels are diminished. This study evaluates whether the exogenous administration of low doses of IGF-1 can induce hepatoprotection in acute carbon tetrachloride (CCl4)-induced liver damage compared to healthy controls (Wt Igf +/+). Additionally, the impact of IGF-1 deficiency on a damaged liver was investigated in mice with a partial deficit of this hormone (Hz Igf1 +/-). METHODS: Three groups of 25 ± 5-week-old healthy male mice (Wt Igf +/+) were included in the protocol: untreated controls (Wt). Controls that received CCl4 (Wt + CCl4) and Wt + CCl4 were treated subcutaneously with IGF-1 (2 µg/100 g body weight/day) for 10 days (Wt + CCl4 + IGF1). In parallel, three IGF-1-deficient mice (Hz Igf1 +/-) groups were studied: untreated Hz, Hz + CCl4, and Hz + CCl4 + IGF-1. Microarray and real-time quantitative polymerase chain reaction (RT-qPCR) analyses, serum aminotransferases levels, liver histology, and malondialdehyde (MDA) levels were assessed at the end of the treatment in all groups. All data represent mean ± SEM. RESULTS: An altered gene coding expression pattern for proteins of the extracellular matrix, fibrosis, and cellular protection were found, as compared to healthy controls, in which IGF-1 therapy normalized in the series including healthy mice. Liver histology showed that Wt + CCl4 + IGF1 mice had less oxidative damage, fibrosis, lymphocytic infiltrate, and cellular changes when compared to the Wt + CCl4. Moreover, there was a correlation between MDA levels and the histological damage score (Pearson's r = 0.858). In the IGF-1-deficient mice series, similar findings were identified, denoting a much more vulnerable hepatic parenchyma. CONCLUSIONS: IGF1 treatment improved the biochemistry, histology, and genetic expression of pro-regenerative and cytoprotective factors in both series (healthy and IGF-1-deficient mice) with acute liver damage, suggesting that low doses of IGF-1, in acute liver damage, could be a feasible therapeutic option.


Subject(s)
Insulin-Like Growth Factor I/deficiency , Insulin-Like Growth Factor I/therapeutic use , Liver Diseases/therapy , Liver/pathology , Animals , Body Weight , Carbon Tetrachloride , Cell Death , Gene Expression Regulation , Insulin-Like Growth Factor I/metabolism , Lipid Peroxidation , Liver/metabolism , Liver Diseases/blood , Liver Diseases/genetics , Male , Mice , Organ Size , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transaminases/blood
5.
Biofactors ; 42(1): 60-79, 2016.
Article in English | MEDLINE | ID: mdl-26891019

ABSTRACT

Insulin-like growth factor 1 (IGF-1) induces multiple cytoprotective effects on every tissue, including the brain. Since the mechanisms by which IGF-1 produces neuroprotection are not fully understood, the aim of this work was to delve into the underlying mechanisms. IGF-1 deficient mice (Hz) were compared with wild type (WT) and Hz mice treated with low doses of IGF-1 (2 µg/100 g body weight/day) for 10 days (Hz + IGF). Gene expression, quantitative PCR, histology, and magnetic resonance imaging were performed in the three groups. IGF-1 deficiency induced increased oxidative damage determined by markers of lipid peroxidation and hypoxia, as well as gene expression of heat shock proteins, antioxidant enzymes, and molecules involved in inflammation, apoptosis, and mitochondrial protection. These changes correlated with edema and learning impairment in Hz mice. IGF-1 therapy improved all these alterations. In conclusion, IGF-1 deficiency is responsible for increased brain oxidative damage, edema, and impaired learning and memory capabilities which are rescued by IGF-1 replacement therapy.


Subject(s)
Brain/metabolism , Edema/metabolism , Inflammation/metabolism , Insulin-Like Growth Factor I/therapeutic use , Animals , Antioxidants/metabolism , Apoptosis/drug effects , Brain/pathology , Brain Injuries/metabolism , Brain Injuries/pathology , Brain Injuries/therapy , Edema/pathology , Edema/therapy , Gene Expression Regulation/drug effects , Heat-Shock Proteins/biosynthesis , Humans , Inflammation/pathology , Inflammation/therapy , Insulin-Like Growth Factor I/deficiency , Insulin-Like Growth Factor I/genetics , Learning/drug effects , Lipid Peroxidation/drug effects , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Neuroprotection/genetics , Oxidative Stress/drug effects
6.
Compr Physiol ; 3(4): 1473-92, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24265236

ABSTRACT

Hepatic stellate cells are resident perisinusoidal cells distributed throughout the liver, with a remarkable range of functions in normal and injured liver. Derived embryologically from septum transversum mesenchyme, their precursors include submesothelial cells that invade the liver parenchyma from the hepatic capsule. In normal adult liver, their most characteristic feature is the presence of cytoplasmic perinuclear droplets that are laden with retinyl (vitamin A) esters. Normal stellate cells display several patterns of intermediate filaments expression (e.g., desmin, vimentin, and/or glial fibrillary acidic protein) suggesting that there are subpopulations within this parental cell type. In the normal liver, stellate cells participate in retinoid storage, vasoregulation through endothelial cell interactions, extracellular matrix homeostasis, drug detoxification, immunotolerance, and possibly the preservation of hepatocyte mass through secretion of mitogens including hepatocyte growth factor. During liver injury, stellate cells activate into alpha smooth muscle actin-expressing contractile myofibroblasts, which contribute to vascular distortion and increased vascular resistance, thereby promoting portal hypertension. Other features of stellate cell activation include mitogen-mediated proliferation, increased fibrogenesis driven by connective tissue growth factor, and transforming growth factor beta 1, amplified inflammation and immunoregulation, and altered matrix degradation. Evolving areas of interest in stellate cell biology seek to understand mechanisms of their clearance during fibrosis resolution by either apoptosis, senescence, or reversion, and their contribution to hepatic stem cell amplification, regeneration, and hepatocellular cancer.


Subject(s)
Fibrosis/pathology , Hepatic Stellate Cells/metabolism , Liver/pathology , Animals , Fibrosis/metabolism , Hepatic Stellate Cells/pathology , Humans , Liver/metabolism
7.
J Transl Med ; 11: 271, 2013 Oct 25.
Article in English | MEDLINE | ID: mdl-24161214

ABSTRACT

BACKGROUND: Bone is one of the major target tissues for Insulin-like Growth Factor I (IGF-I). Low doses of IGF-I were able to improve liver-associated osteopenia. In the present work, a model of partial IGF-I deficiency was used in order to provide insight into the mechanisms of the beneficial actions of IGF-I replacement therapy in bone. METHODS: Several proteins involved in osteoblastic/osteocyte and osteoclastic differentiation and activity were studied in the three experimental groups: control (CO) group (wild type mice, Igf+/+, n=10), heterozygous Igf+/- group with partial IGF-I deficiency (Hz, n=10), and heterozygous Igf+/- mice treated with IGF-I for 10 days (Hz+IGF-I, n=10). RESULTS: Data in this paper confirm that the simple partial IGF-I deficiency is responsible for osteopenia, determined by densitometry and histopathology. These findings are associated with a reduced gene expression of osteoprotegerin, sclerostin, calcitonin receptor (CTR), insulin-like growth factor binding protein 5 and RUNX2. IGF-I replacement therapy normalized CTR gene expression and reduced markers of osteoclastic activity. CONCLUSIONS: Low doses of IGF-I constituted a real replacement therapy that normalized IGF-I serum levels improving the expression of most of these proteins closely involved in bone-forming, and reducing bone resorption by mechanisms related to osteoprotegerin, RANKL and PTH receptor.


Subject(s)
Biomarkers/metabolism , Bone Resorption , Insulin-Like Growth Factor I/metabolism , Osteoblasts/metabolism , Osteoprotegerin/metabolism , RANK Ligand/metabolism , Animals , Insulin-Like Growth Factor I/administration & dosage , Mice , Mice, Transgenic , Reverse Transcriptase Polymerase Chain Reaction
8.
Hepatology ; 57(1): 339-50, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22961591

ABSTRACT

UNLABELLED: We have developed a novel model for depleting mouse hepatic stellate cells (HSCs) that has allowed us to clarify their contributions to hepatic injury and fibrosis. Transgenic (Tg) mice expressing the herpes simplex virus thymidine kinase gene (HSV-Tk) driven by the mouse GFAP promoter were used to render proliferating HSCs susceptible to killing in response to ganciclovir (GCV). Effects of GCV were explored in primary HSCs and in vivo. Panlobular damage was provoked to maximize HSC depletion by combining CCl(4) (centrilobular injury) with allyl alcohol (AA) (periportal injury), as well as in a bile duct ligation (BDL) model. Cell depletion in situ was quantified using dual immunofluorescence (IF) for desmin and GFAP. In primary HSCs isolated from both untreated wild-type (WT) and Tg mice, GCV induced cell death in ≈ 50% of HSCs from Tg, but not WT, mice. In TG mice treated with CCl(4) +AA+GCV, there was a significant decrease in GFAP and desmin-positive cells, compared to WT mice (≈ 65% reduction; P < 0.01), which was accompanied by a decrease in the expression of HSC-activation markers (alpha smooth muscle actin, beta platelet-derived growth factor receptor, and collagen I). Similar results were observed after BDL. Associated with HSC depletion in both fibrosis models, there was marked attenuation of fibrosis and liver injury, as indicated by Sirius Red/Fast Green, hematoxylin and eosin quantification, and serum alanine/aspartate aminotransferase. Hepatic expression of interleukin-10 and interferon-gamma was increased after HSC depletion. No toxicity of GCV in either WT or Tg mice accounted for the differences in injury. CONCLUSION: Activated HSCs significantly amplify the response to liver injury, further expanding this cell type's repertoire in orchestrating hepatic injury and repair.


Subject(s)
Chemical and Drug Induced Liver Injury/etiology , Hepatic Stellate Cells/physiology , Animals , Apoptosis , Carbon Tetrachloride , Cell Line , Chemical and Drug Induced Liver Injury/pathology , Cytokines/metabolism , Fibrosis , Ganciclovir , Glial Fibrillary Acidic Protein , Liver/immunology , Liver/pathology , Male , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Promoter Regions, Genetic , Propanols , Thymidine Kinase/genetics , Viral Proteins/genetics
9.
J Transl Med ; 10: 224, 2012 Nov 14.
Article in English | MEDLINE | ID: mdl-23148873

ABSTRACT

Insulin-like growth factor I (IGF-I) is a polypeptide hormone produced mainly by the liver in response to the endocrine GH stimulus, but it is also secreted by multiple tissues for autocrine/paracrine purposes. IGF-I is partly responsible for systemic GH activities although it possesses a wide number of own properties (anabolic, antioxidant, anti-inflammatory and cytoprotective actions). IGF-I is a closely regulated hormone. Consequently, its logical therapeutical applications seems to be limited to restore physiological circulating levels in order to recover the clinical consequences of IGF-I deficiency, conditions where, despite continuous discrepancies, IGF-I treatment has never been related to oncogenesis. Currently the best characterized conditions of IGF-I deficiency are Laron Syndrome, in children; liver cirrhosis, in adults; aging including age-related-cardiovascular and neurological diseases; and more recently, intrauterine growth restriction. The aim of this review is to summarize the increasing list of roles of IGF-I, both in physiological and pathological conditions, underlying that its potential therapeutical options seem to be limited to those proven states of local or systemic IGF-I deficiency as a replacement treatment, rather than increasing its level upper the normal range.


Subject(s)
Disease , Insulin-Like Growth Factor I/deficiency , Animals , Growth and Development , Humans , Insulin-Like Growth Factor I/chemistry , Insulin-Like Growth Factor I/metabolism , Organ Specificity
10.
Mol Cancer Res ; 10(9): 1216-27, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22859706

ABSTRACT

Alternative splicing of the Krüppel-like factor 6 (KLF6) tumor suppressor into an antagonistic splice variant 1 (SV1) is a pathogenic event in several cancers including hepatocellular carcinoma (HCC) because elevated SV1 is associated with increased tumor metastasis and mortality. Ras activation is one factor that can enhance KLF6 splicing in cancer cells, however pathways driving KLF6 splicing are unknown. Splice site selection is regulated by splice factors that include serine/arginine-rich (SR) proteins such as SRSF1 (ASF-SF2), which in turn is controlled by phosphoinositide 3-kinase (PI3K)/Akt and the mitogen-activated protein kinase (MAPK) signaling pathway. Because signaling pathways downstream of the liver mitogen hepatocyte growth factor (HGF) include Akt, we explored whether HGF induces KLF6 alternative splicing. In HepG2 cells, HGF (25 ng/mL) significantly increases the ratio of SV1/KLF6 full by 40% through phosphorylation of Akt and subsequent downregulation of two splicing regulators, SRSF3 (SRp20) and SRSF1. Decreased SRSF3 levels regulate SRSF1 levels by alternative splicing associated with the nonsense-mediated mRNA decay pathway (AS-NMD), which stimulates cell growth by decreasing p21 levels. Enhanced cell replication through increased KLF6 alternative splicing is a novel growth-promoting pathway of HGF that could contribute to the molecule's mitogenic activity in physiologic liver growth and hepatocellular carcinoma.


Subject(s)
Alternative Splicing , Carcinoma, Hepatocellular/metabolism , Hepatocyte Growth Factor/metabolism , Kruppel-Like Transcription Factors/metabolism , Liver Neoplasms/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/metabolism , RNA-Binding Proteins/metabolism , Base Sequence , Carcinoma, Hepatocellular/genetics , Cell Proliferation , Down-Regulation , Gene Expression Regulation, Neoplastic , Hep G2 Cells , Humans , Kruppel-Like Factor 6 , Kruppel-Like Transcription Factors/genetics , Liver Neoplasms/genetics , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Molecular Sequence Data , Neoplasm Metastasis , Nonsense Mediated mRNA Decay , Nuclear Proteins/genetics , Phosphorylation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Neoplasm/genetics , RNA-Binding Proteins/genetics , Sequence Analysis, DNA , Serine-Arginine Splicing Factors , Signal Transduction
11.
J Transl Med ; 9: 103, 2011 Jul 06.
Article in English | MEDLINE | ID: mdl-21733157

ABSTRACT

BACKGROUND: GH and IGFs serum levels decline with age. Age-related changes appear to be associated to decreases in these anabolic hormones. We have previously demonstrated that IGF-I replacement therapy improves insulin resistance, lipid metabolism and reduces oxidative damage (in brain and liver) in aging rats. Using the same experimental model, the aim of this work was to study whether the exogenous administration of IGF-II, at low doses, acts analogous to IGF-I in aging rats. METHODS: Three experimental groups were included in this study: young healthy controls (yCO, 17 weeks old); untreated old rats (O, 103 weeks old); and aging rats treated with IGF-II (O+IGF-II, 2 µg * 100 g body weight⁻¹ * day⁻¹) for 30 days. Analytical parameters were determined in serum by routine laboratory methods using an autoanalyzer (Cobas Mira; Roche Diagnostic System, Basel, Switzerland). Serum levels of hormones (testosterone, IGF-I and insulin) were assessed by RIA. Serum Total Antioxidant Status was evaluated using a colorimetric assay. Mitochondrial membrane potential was evaluated using rhodamine 123 dye (adding different substrates to determine the different states). ATP synthesis in isolated mitochondria was determined by an enzymatic method. RESULTS: Compared with young controls, untreated old rats showed a reduction of IGF-I and testosterone levels with a decrease of serum total antioxidant status (TAS). IGF-II therapy improved serum antioxidant capability without modifying testosterone and IGF-I circulating concentrations. In addition, IGF-II treatment reduced oxidative damage in brain and liver, improving antioxidant enzyme activities and mitochondrial function. IGF-II was also able to reduce cholesterol and triglycerides levels increasing free fatty acids concentrations. CONCLUSIONS: We demonstrate that low doses of IGF-II induce hepatoprotective, neuroprotective and metabolic effects, improving mitochondrial function, without affecting testosterone and IGF-I levels.


Subject(s)
Aging/drug effects , Insulin-Like Growth Factor II/administration & dosage , Insulin-Like Growth Factor II/pharmacology , Liver/drug effects , Nervous System/drug effects , Protective Agents/pharmacology , Aging/metabolism , Animals , Antioxidants/metabolism , Brain/drug effects , Brain/enzymology , Dose-Response Relationship, Drug , Glucose/metabolism , Hormones/metabolism , Humans , Insulin/blood , Insulin-Like Growth Factor I/metabolism , Lipid Metabolism/drug effects , Lipid Peroxidation/drug effects , Liver/enzymology , Male , Malondialdehyde/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Nervous System/metabolism , Oxidative Stress/drug effects , Protein Carbonylation/drug effects , Rats , Rats, Wistar , Testosterone/blood
12.
J Transl Med ; 9: 123, 2011 Jul 28.
Article in English | MEDLINE | ID: mdl-21798010

ABSTRACT

BACKGROUND: Serum IGF-I and IGF-II levels decline with age. IGF-I replacement therapy reduces the impact of age in rats. We have recently reported that IGF-II is able to act, in part, as an analogous of IGF-I in aging rats reducing oxidative damage in brain and liver associated with a normalization of antioxidant enzyme activities. Since mitochondria seem to be the most important cellular target of IGF-I, the aim of this work was to investigate whether the cytoprotective actions of IGF-II therapy are mediated by mitochondrial protection. METHODS: Three groups of rats were included in the experimental protocol young controls (17 weeks old); untreated old rats (103 weeks old); and aging rats (103 weeks old) treated with IGF-II (2 µg/100 g body weight and day) for 30 days. RESULTS: Compared with young controls, untreated old rats showed an increase of oxidative damage in isolated mitochondria with a dysfunction characterized by: reduction of mitochondrial membrane potential (MMP) and ATP synthesis and increase of intramitochondrial free radicals production and proton leak rates. In addition, in untreated old rats mitochondrial respiration was not blocked by atractyloside. In accordance, old rats showed an overexpression of the active fragment of caspases 3 and 9 in liver homogenates. IGF-II therapy corrected all of these parameters of mitochondrial dysfunction and reduced activation of caspases. CONCLUSIONS: The cytoprotective effects of IGF-II are related to mitochondrial protection leading to increased ATP production reducing free radical generation, oxidative damage and apoptosis.


Subject(s)
Aging/drug effects , Aging/metabolism , Insulin-Like Growth Factor II/pharmacology , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , ATP Synthetase Complexes/metabolism , Animals , Antioxidants/metabolism , Atractyloside/pharmacology , Caspase 3/metabolism , Caspase 9 , Electron Transport Complex IV/metabolism , Humans , Hydrogen Peroxide/metabolism , Insulin-Like Growth Factor II/administration & dosage , Male , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Liver/enzymology , Oxidative Stress/drug effects , Oxygen Consumption/drug effects , Protons , Rats , Rats, Wistar
13.
World J Gastroenterol ; 14(17): 2731-9, 2008 May 07.
Article in English | MEDLINE | ID: mdl-18461658

ABSTRACT

AIM: To characterize the mitochondrial dysfunction in experimental cirrhosis and to study whether insulin-like growth factor-I (IGF- I) therapy (4 wk) is able to induce beneficial effects on damaged mitochondria leading to cellular protection. METHODS: Wistar rats were divided into three groups: Control group, untreated cirrhotic rats and cirrhotic rats treated with IGF- I treatment (2 microg/100 g bw/d). Mitochondrial function was analyzed by flow cytometry in isolated hepatic mitochondria, caspase 3 activation was assessed by Western blot and apoptosis by TUNEL in the three experimental groups. RESULTS: Untreated cirrhotic rats showed a mitochondrial dysfunction characterized by a significant reduction of mitochondrial membrane potential (in status 4 and 3); an increase of intramitochondrial reactive oxigen species (ROS) generation and a significant reduction of ATPase activity. IGF- I therapy normalized mitochondrial function by increasing the membrane potential and ATPase activity and reducing the intramitochondrial free radical production. Activity of the electron transport complexes I and III was increased in both cirrhotic groups. In addition, untreated cirrhotic rats showed an increase of caspase 3 activation and apoptosis. IGF- I therapy reduced the expression of the active peptide of caspase 3 and resulted in reduced apoptosis. CONCLUSION: These results show that IGF- I exerts a mitochondrial protection in experimental cirrhosis leading to reduced apoptosis and increased ATP production.


Subject(s)
Insulin-Like Growth Factor I/pharmacology , Liver Cirrhosis, Experimental/prevention & control , Mitochondria, Liver/drug effects , Animals , Apoptosis/drug effects , Blotting, Western , Carbon Tetrachloride , Caspase 3/metabolism , Electron Transport Chain Complex Proteins/metabolism , Flow Cytometry , Free Radicals/metabolism , Humans , In Situ Nick-End Labeling , Liver Cirrhosis, Experimental/chemically induced , Liver Cirrhosis, Experimental/metabolism , Liver Cirrhosis, Experimental/pathology , Male , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Liver/metabolism , Mitochondria, Liver/pathology , Mitochondrial Proton-Translocating ATPases/metabolism , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Recombinant Proteins/pharmacology
14.
Endocrinology ; 149(5): 2620-7, 2008 May.
Article in English | MEDLINE | ID: mdl-18276748

ABSTRACT

Serum IGF-I levels decline with age. We have recently reported that in aging rats the exogenous administration of IGF-I restores IGF-I circulating levels and age related-changes, improving glucose and lipid metabolisms, increasing testosterone levels and serum total antioxidant capability, and reducing oxidative damage in the brain and liver associated with a normalization of antioxidant enzyme activities. Understanding that mitochondria are one of the most important cellular targets of IGF-I, the aims of this study were to characterize mitochondrial dysfunction and study the effect of IGF-I therapy on mitochondria, leading to cellular protection in the following experimental groups: young controls, untreated old rats, and aging rats treated with IGF-I. Compared with young controls, untreated aging rats showed an increase of oxidative damage in isolated mitochondria with a mitochondrial dysfunction characterized by: depletion of membrane potential with increased proton leak rates and intramitochondrial free radical production, and a significant reduction of ATPase and complex IV activities. In addition, mitochondrial respiration from untreated aging rats was atractyloside insensitive, suggesting that the adenine nucleotide translocator was uncoupled. The adenine nucleotide translocator has been shown to be one of the most sensitive locations for pore opening. Accordingly, untreated aging rats showed a significant overexpression of the active fragment of caspases 3 and 9. IGF-I therapy corrected these parameters of mitochondrial dysfunction and reduced caspase activation. In conclusion, these results show that the cytoprotective effect of IGF-I is closely related to a mitochondrial protection, leading to reduce free radical production, oxidative damage, and apoptosis, and to increased ATP production.


Subject(s)
Aging/drug effects , Cytoprotection/drug effects , Insulin-Like Growth Factor I/pharmacology , Mitochondria, Liver/drug effects , Adenosine Triphosphatases/metabolism , Animals , Antioxidants/metabolism , Caspases/metabolism , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Electron Transport Complex IV/metabolism , Hepatocytes/drug effects , Male , Matrix Metalloproteinases/metabolism , Mitochondria, Liver/metabolism , Mitochondria, Liver/physiology , Oxidative Stress/drug effects , Oxygen Consumption/drug effects , Proton Pumps/metabolism , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...