Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Med Sci Monit Basic Res ; 25: 229-237, 2019 Nov 07.
Article in English | MEDLINE | ID: mdl-31695019

ABSTRACT

BACKGROUND Progranulin is an adipokine, encoded by the progranulin (GRN) gene. Progranulin is expressed in atherosclerosis, but its effects in cardiac ischemia and reperfusion injury are unknown. Therefore, this study aimed to investigate the effects of progranulin in a rat model of acute myocardial ischemia/reperfusion (MI/R) injury in vivo. MATERIAL AND METHODS The model of acute MI/R injury was established in male Wistar rats by ligation of the left anterior descending (LAD) coronary artery for 30 minutes and reperfusion for 60 minutes. Before modeling, one group was treated with progranulin (0.03 µg/kg), and one group was treated with the P13K/Akt inhibitor, LY294002 (3 mg/kg). Left ventricular function (LV) was monitored, including the LV systolic pressure (LVSP), LV end-diastolic pressure (LVEDP), and changes in LV pressure. At the end of the study, blood and myocardial tissue were examined. Cardiac biochemical markers, histopathology, gene expression, and apoptosis were analyzed. RESULTS Progranulin improved cardiac function following acute MI/R injury and significantly improved recovery of cardiac contractility and LVSP. Progranulin significantly reduced myocyte apoptosis, inflammation, and tissue edema, and was highly expressed in cardiac tissue following MI/R injury. The cardioprotective effect of progranulin was reduced by blocking the P13K/Akt signaling pathway. CONCLUSIONS In the rat model of acute MI/R injury, progranulin had a protective effect on cardiac function and morphology, associated with activation of the P13K/Akt signaling pathway. The mechanisms of the anti-apoptotic, anti-inflammatory, and inotropic effects of progranulin in the setting of acute MI/R injury require further in vivo studies.


Subject(s)
Myocardial Reperfusion Injury/drug therapy , Progranulins/pharmacology , Adipokines/pharmacology , Animals , Apoptosis/drug effects , Atherosclerosis/drug therapy , Chromones/pharmacology , Disease Models, Animal , Male , Morpholines/pharmacology , Myocardial Ischemia/physiopathology , Myocardium/pathology , Myocytes, Cardiac/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Progranulins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Reperfusion Injury/drug therapy , Signal Transduction/drug effects
2.
Cell Stress Chaperones ; 19(2): 173-81, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23801458

ABSTRACT

Peroxiredoxins, a group of antioxidant protein enzymes (PRDX1 to 6), are reported as antiatherogenic factors in animals; however, human studies are lacking. The present work aims to provide baseline data regarding the phenotype of PRDX1, 2, 4, and 6 in diabetic patients with peripheral atherosclerosis disease (PAD) and their relation to endothelial dysfunction (ED) and disease severity. Plasma levels of PRDX1, 2, 4, and 6 and markers of endothelial dysfunction (ICAM-1 and VCAM-1) were measured using ELISA in 55 type 2 diabetic patients having PAD and 25 healthy subjects. Ankle-brachial index (ABI), body mass index (BMI), triglycerides (TG), total cholesterol, HbA1c, and insulin resistance (HOMA IR) were measured. PRDX1, 2, 4, and 6 levels were significantly higher in patients compared to controls (PRDX1 21.9 ± 5.71 vs 16.8 ± 3.9 ng/ml, P < 0.001, PRDX2 36.5 ± 14.83 vs 20.4 ± 8.61 ng/ml, P < 0.001, PRDX4 3,840 ± 1,440 vs 2,696 ± 1,972 pg/ml, P < 0.005, PRDX6 311 ± 110 vs 287.9 ± 114 pg/ml, P < 0.05). PRDX1 and PRDX4 correlated negatively with ABI (r = -0.273, P < 0.05 and r = -0.28, P < 0.05, respectively), while PRDX1 and PRDX2 correlated positively with HOMA/IR and TG (r = 0.276, P < 0.01 and r = 0.295, P < 0.01, respectively). ICAM-1 was associated with PRDX2 and log PRDX6 (r = 0.345, P = 0.0037 and r = 0.344, P = 0.0038). Our results provide strong links among PRDXs, ED, and severity of PAD in diabetic patients which warrants further evaluation to clarify whether high circulating levels of PRDXs are a consequence of chronic atherosclerotic disease or a predisposing factor for later cardiovascular events.


Subject(s)
Atherosclerosis/physiopathology , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/physiopathology , Endothelium, Vascular/physiopathology , Peripheral Arterial Disease/blood , Peripheral Arterial Disease/physiopathology , Peroxiredoxins/blood , Ankle Brachial Index , Atherosclerosis/blood , Atherosclerosis/complications , Atherosclerosis/pathology , Biomarkers/blood , Case-Control Studies , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/pathology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Female , Humans , Male , Middle Aged , Peripheral Arterial Disease/complications , Peripheral Arterial Disease/pathology
3.
Exp Gerontol ; 51: 1-7, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24373843

ABSTRACT

Hypercholesterolemia (HC) and aging combine to increase the incidence of cerebrovascular disease through oxidative stress. Our investigation examined the effects of diet-induced hypercholesterolemia (2% for 8weeks) on the extent of brain injury in response to global cerebral ischemia/reperfusion (GCI/R) and the neuroprotective potentials of rosiglitazone in relation to oxidative stress. HC exacerbated the decline in the brain levels of GSH and the increase in MPO, proinflammatory markers and hippocampal lesions in response to GCI/R. HC rats receiving rosiglitazone, PPAR-γ agonist, demonstrated preservation of cell viability of CA1 hippocampal region and attenuation of brain edema. They also showed elevated levels of GSH and low levels of the other parameters similar to non-HC rats subjected to GCI/R. In vitro, rosiglitazone dose-dependently inhibited ROS generation by neutrophils. The results suggest exacerbation of brain lesions by HC in response to GCI/R. The neuroprotective therapeutic potentials of rosiglitazone are comparable to non-HC animals. Mechanisms of protection are possibly due to anti-oxidant, anti-inflammatory effects and scavenging properties of rosiglitazone. These results add to the beneficial therapeutic effects of rosiglitazone and its significance for age-associated diseases including hypercholesterolemia.


Subject(s)
Brain Ischemia/drug therapy , Hypercholesterolemia/drug therapy , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , PPAR gamma/agonists , Reperfusion Injury/prevention & control , Thiazolidinediones/pharmacology , Animals , Brain Edema/prevention & control , Dose-Response Relationship, Drug , Glutathione/drug effects , Glutathione/metabolism , Interleukin-6/metabolism , Lipid Peroxidation/drug effects , Male , Peroxidase/drug effects , Peroxidase/metabolism , Rats , Rats, Wistar , Rosiglitazone , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/metabolism
4.
Cardiovasc Hematol Agents Med Chem ; 11(3): 179-86, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23952292

ABSTRACT

Apoptosis signal-regulating kinase 1 (ASK1) is among the signaling events that lead to postischemic cell death. Inhibition of ASK1 pathway protected hearts from ischemic damage. The present study evaluated the renal protective effects of NQDI 1, an inhibitor of ASK1, in an animal model of acute ischemic renal failure. Male Wistar rats were subjected to right nephrectomy and clamping of left renal pedicle for 45 min, or sham operation. The administration of NQDI 1 attenuated renal dysfunction and histological changes characteristic for renal ischemia/reperfusion injury (IRI). Apoptosis of renal tissues, as detected by TUNEL staining, was also reduced together with p53 protein expression, and renal levels of MDA and SOD with NQDI 1 administration and BCL2 was up regulated. In conclusion, inhibition of ASK1 is of therapeutic potential against acute ischemic renal injury. Its protective effects are mediated via inhibition of apoptosis and oxidative stress.


Subject(s)
Aporphines/pharmacology , Enzyme Inhibitors/pharmacology , Kidney/drug effects , MAP Kinase Kinase Kinase 5/antagonists & inhibitors , Oxidative Stress/drug effects , Quinolines/pharmacology , Reperfusion Injury/enzymology , Animals , Aporphines/chemistry , Cell Death/drug effects , Disease Models, Animal , Kidney/enzymology , Kidney/physiopathology , Male , Quinolines/chemistry , Rats , Rats, Wistar
5.
World J Gastroenterol ; 18(18): 2188-96, 2012 May 14.
Article in English | MEDLINE | ID: mdl-22611311

ABSTRACT

AIM: To investigate the effect of administration of agmatine (AGM) on gastric protection against ischemia reperfusion (I/R) injury. METHODS: Three groups of rats (6/group); sham, gastric I/R injury, and gastric I/R + AGM (100 mg/kg, i.p. given 15 min prior to gastric ischemia) were recruited. Gastric injury was conducted by ligating celiac artery for 30 min and reperfusion for another 30 min. Gastric tissues were histologically studied and immunostained with angiopoietin 1 (Ang-1) and Ang-2. Vascular endothelial growth factor (VEGF) and monocyte chemoattractant protein-1 (MCP-1) were measured in gastric tissue homogenate. To assess whether AKt/phosphatidyl inositol-3-kinase (PI3K) mediated the effect of AGM, an additional group was pretreated with Wortmannin (WM) (inhibitor of Akt/PI3K, 15 µg/kg, i.p.), prior to ischemic injury and AGM treatment, and examined histologically and immunostained. Another set of experiments was run to study vascular permeability of the stomach using Evan's blue dye. RESULTS: AGM markedly reduced Evan's blue dye extravasation (3.58 ± 0.975 µg/stomach vs 1.175 ± 0.374 µg/stomach, P < 0.05), VEGF (36.87 ± 2.71 pg/100 mg protein vs 48.4 ± 6.53 pg/100 mg protein, P < 0.05) and MCP-1 tissue level (29.5 ± 7 pg/100 mg protein vs 41.17 ± 10.4 pg/100 mg protein, P < 0.01). It preserved gastric histology and reduced congestion. Ang-1 and Ang-2 immunostaining were reduced in stomach sections of AGM-treated animals. The administration of WM abolished the protective effects of AGM and extensive hemorrhage and ulcerations were seen. CONCLUSION: AGM protects the stomach against I/R injury by reducing vascular permeability and inflammation. This protection is possibly mediated by Akt/PI3K.


Subject(s)
Agmatine/pharmacology , Capillary Permeability/drug effects , Protective Agents/pharmacology , Reperfusion Injury/prevention & control , Stomach/blood supply , Stomach/drug effects , Androstadienes/pharmacology , Angiopoietin-1/metabolism , Angiopoietin-2/metabolism , Animals , Chemokine CCL2/metabolism , Cytoprotection , Disease Models, Animal , Down-Regulation , Gastric Mucosa/metabolism , Immunohistochemistry , Male , Phosphatidylinositol 3-Kinase/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Stomach/pathology , Vascular Endothelial Growth Factor A/metabolism , Wortmannin
6.
Shock ; 37(6): 639-44, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22592635

ABSTRACT

Renal ischemia-reperfusion injury (I/R) is the most common cause of acute renal failure. It is partially mediated by thrombin as it is attenuated by thrombin inhibition or deletion of its receptor protease-activated receptor 1 (PAR1). However, the role of PAR1 in renal I/R injury needs to be further elucidated. The present study investigated the effect of PAR1 antagonist, SCH79797 (SCH), on renal protection and downstream effectors involved. Male Wistar rats were pretreated with SCH (25 µg/kg i.p.) or vehicle, 15 min before 45 min of clamping of left renal pedicle after right nephrectomy. To investigate the involvement of phosphatidylinositol 3-kinase (PI3K)/Akt, a group of rats was subjected to pretreatment with an inhibitor of PI3K/Akt (LY 29004, 3 mg/kg i.p.) before renal ischemia and SCH treatment. A sham-operated group served as control and received saline. All rats were killed 24 h after reperfusion or sham operation, and blood samples collected and kidney tissues processed either for immunostaining and histological assessment or for biochemical analysis. SCH79797 markedly attenuated kidney damage histologically and by improving serum creatinine. Both plasma and protein expression of P selectin were markedly reduced as well as neutrophil infiltration, cytokine-induced neutrophil chemoattractant 1, and tumor necrosis factor α. These protective effects of blocking PAR1 receptor were abolished by preadministration of LY29004. These results suggest that PAR1 mediates renal I/R injury and that blocking PAR1 using SCH limits renal injury by an anti-inflammatory effect possibly signaling via PI3K/Akt.


Subject(s)
Kidney/blood supply , Protease Inhibitors/pharmacology , Pyrroles/pharmacology , Quinazolines/pharmacology , Receptor, PAR-1/antagonists & inhibitors , Reperfusion Injury/metabolism , Animals , Chemokine CXCL1/metabolism , Creatinine/blood , Kidney/metabolism , Kidney/pathology , Male , Neutrophil Infiltration , P-Selectin/blood , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Reperfusion Injury/prevention & control , Tumor Necrosis Factor-alpha/metabolism
7.
J Gastroenterol Hepatol ; 22(11): 1791-9, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17914952

ABSTRACT

BACKGROUND AND AIM: Gherlin, an endogenous ligand for the growth hormone secretagogue receptor (GHS-R), is produced by stomach cells. It regulates food intake, gastric secretion and motility. However, its role as a protective agent in gastric ischemia/reperfusion (I/R) injury has not yet been investigated. Therefore, the objectives of the present study were to: (i) test the in vivo effect of peripherally administered ghrelin on gastric I/R-induced lesions in rats; and (ii) investigate in vitro the effect of ghrelin on reactive oxygen species (ROS) production by human polymorphoneuclear (PMN) cells. METHODS: The present study was carried out on three groups of rats (six per group): control (sham-operated), I/R (clamping of celiac artery for 30 min and reperfusion for 1 h), and I/R + ghrelin (200 ng/kg i.v., 15 min before ischemia and before reperfusion, respectively). Histological assessment of hematoxylin and eosin stained sections was performed and immunostaining with inducible nitric oxide (iNOS) antibody were performed on a gastric paraffin embedded section. Oxidative stress markers thiobarbituric acid reactive substance (TBARS) and glutathione (GSH) were measured in gastric tissue homogenates. Serum lactic acid dehydrogenase (LDH) was determined. Tumor necrosis factor-alpha (TNF-alpha) was assayed in gastric tissue homogenate. Gastric permeability was assessed calorimetrically using Evans blue dye. In vitro studies were carried out on isolated human PMN cells incubated with ghrelin and tested for ROS generation as measured by chemiluminecence (CL). RESULTS: Peripheral administration of ghrelin attenuated gastric injury by reducing ulceration, tissue congestion, cellular infiltration and vascular permeability. Serum level of LDH and tissue content of TNF-alpha were markedly reduced. A decrement in TBARS and an increment in GSH were observed. Ghrelin treatment attenuated iNOS protein expression which was upregulated by gastric ischemic injury. In vitro studies showed for the first time that ghrelin inhibited ROS generation by human PMN in a dose-dependent manner. CONCLUSIONS: These results provide evidence that peripherally administered ghrelin protects against gastric I/R injury. We also demonstrated that this protection is possibly accomplished through the antioxidant activity of ghrelin observed in vivo and in vitro.


Subject(s)
Antioxidants/metabolism , Ghrelin/metabolism , Leukocytes, Mononuclear/metabolism , Oxidative Stress , Reperfusion Injury/prevention & control , Stomach/drug effects , Animals , Antioxidants/administration & dosage , Capillary Permeability/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Gastric Mucosa/metabolism , Ghrelin/administration & dosage , Glutathione/metabolism , Humans , In Vitro Techniques , Injections, Intravenous , L-Lactate Dehydrogenase/blood , Leukocytes, Mononuclear/drug effects , Male , Nitric Oxide Synthase Type II/metabolism , Oxidative Stress/drug effects , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Stomach/blood supply , Stomach/pathology , Thiobarbituric Acid Reactive Substances/metabolism , Time Factors , Tumor Necrosis Factor-alpha/blood
8.
Eur J Pharmacol ; 554(1): 69-77, 2007 Jan 05.
Article in English | MEDLINE | ID: mdl-17112501

ABSTRACT

Ulcerative colitis is a chronically recurrent inflammatory bowel disease of unknown origin. The present study examined the effect of NF-kappaB inhibitor and antioxidant, pyrrolidinedithiocarbamate (PDTC) on experimental ulcerative colitis in rats. Animals were randomly divided into 4 groups, each consisting of 6 animals; normal control group, acetic acid group, PDTC-treated group and sulfasalazine-treated group as a positive control group. Induction of colitis by intracolonic administration of 3% acetic acid produced severe macroscopic inflammation in the colon 24 h after acetic acid administration as assessed by the colonic damage score. Microscopically, colonic tissues showed ulceration, oedema and inflammatory cells infiltration. Biochemical studies revealed increased serum levels of lactate dehydrogenase (LDH), and nitrite/nitrate and colonic concentrations of tumor necrosis factor-alpha (TNF-alpha) and the neutrophil infiltration index, myeloperoxidase (MPO). Oxidative stress was indicated by elevated lipid peroxides formation and depleted reduced glutathione concentrations (GSH) in colonic tissues. Immunohistochemical studies of colonic sections revealed upregulation of inducible nitric oxide synthase (iNOS). Pretreatment with PDTC at a dose of (200 mg/kg/day, i.p.), three days before induction of colitis decreased serum LDH, nitrite/nitrate and TNF-alpha levels, colonic concentrations of MPO and lipid peroxides while increased colonic GSH concentration. Moreover, PDTC pretreatment attenuated colonic iNOS expression. Finally, histopathological changes were nearly restored by PDTC pretreatment. The findings of the present study provide evidence that PDTC may be beneficial in patients with inflammatory bowel disease.


Subject(s)
Colitis, Ulcerative/drug therapy , Proline/analogs & derivatives , Thiocarbamates/therapeutic use , Acetic Acid/toxicity , Animals , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/pathology , Colon/chemistry , Glutathione/analysis , L-Lactate Dehydrogenase/blood , Lipid Peroxides/analysis , Male , Nitrates/blood , Nitric Oxide Synthase Type II/analysis , Nitrites/blood , Peroxidase/metabolism , Proline/therapeutic use , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/analysis
9.
Can J Physiol Pharmacol ; 83(6): 483-92, 2005 Jun.
Article in English | MEDLINE | ID: mdl-16049548

ABSTRACT

Pyrrolidinedithiocarbamate (PDTC) is a potent antioxidant and an inhibitor of nuclear factor-kappaB (NF-kappaB). The present study examined the impact of PDTC preconditioning on gastric protection in response to ischemia-reperfusion (I/R) injury to the rat stomach. Male Wistar rats were recruited and divided into 3 groups (n = 7). One group was subjected to gastric ischemia for 30 min and reperfusion for 1 hour. The second group of rats was preconditioned with PDTC (200 mg/kg body mass i.v.) 15 min prior to ischemia and before reperfusion. The third group of rats was sham-operated and served as the control group. Gastric I/R injury increased serum lactate dehydrogenase level, vascular permeability of gastric mucosa (as indicated by Evans blue dye extravasation) and gastric content of inflammatory cytokine; tumor necrosis factor-alpha (TNF-alpha). Moreover, oxidative stress was increased as indicated by elevated lipid peroxides formation (measured as thiobarbituric acid reactive substances) and depleted reduced glutathione in gastric tissues. NF-kappaB translocation was also detected by electrophoretic mobility shift assay. Microscopically, gastric tissues subjected to I/R injury showed ulceration, hemorrhages, and neutrophil infiltration. Immunohistochemical studies of gastric sections revealed increased expression of p53 and Bcl-2 proteins. PDTC pretreatment reduced Evans blue extravasation, serum lactate dehydrogenase levels, gastric TNF-alpha levels, and thiobarbituric acid reactive substances content, and increased gastric glutathione content. Moreover, PDTC pretreatment abolished p53 expression and inhibited NF-kappaB translocation. Finally, histopathological changes were nearly restored by PDTC pretreatment. These results clearly demonstrate that NF-kappaB activation and pro-apoptotic protein p53 induction are involved in gastric I/R injury. PDTC protects against gastric I/R injury by an antioxidant, NF-kappaB inhibition, and by reduction of pro-apoptotic protein p53 expression, which seems to be downstream to NF-kappaB, thus promoting cell survival.


Subject(s)
Antioxidants/therapeutic use , NF-kappa B/antagonists & inhibitors , Pyrrolidines/therapeutic use , Reperfusion Injury/prevention & control , Stomach/blood supply , Thiocarbamates/therapeutic use , Animals , Gastric Mucosa/blood supply , Gastric Mucosa/drug effects , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Glutathione/metabolism , Immunohistochemistry , Lipid Peroxides/biosynthesis , Male , Oxidative Stress/drug effects , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Rats , Rats, Wistar , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Stomach/drug effects , Stomach/pathology , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Protein p53/biosynthesis
10.
J Am Soc Nephrol ; 11(11): 2027-2035, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11053478

ABSTRACT

Biochemical evidence suggests that insulin-like growth factor I (IGF-I) may play an important role as a mediator of kidney growth. In the present study, an IGF-I receptor antagonist (JB3) was synthesized, and its effect on the renal growth that follows the induction of diabetes or unilateral nephrectomy (UNx) was examined. JB3 was generated by solid phase peptide synthesis. Its activity as an IGF-I antagonist was confirmed in an opossum kidney cell line from its inhibitory effect on the increase in thymidine incorporation into DNA induced by recombinant human IGF-I. Male Wistar rats were anesthetized with halothane and subjected to either the induction of diabetes by streptozotocin (intravenous 60 mg/kg) for 4 d (control animals received citrate buffer) or UNx for 11 d (control animals were sham operated). JB3 was delivered by subcutaneous infusion using an osmotic minipump implanted 3 d before the induction of diabetes or UNx. Kidney wet weight, DNA, and protein all were significantly higher 4 d after the induction of diabetes (24%) or 11 d after UNx (55%). Dose-response studies (1 to 30 microg/kg per day) showed JB3 administration to inhibit the increase in kidney growth in both diabetic and UNx rats. The increase in kidney wet weight, DNA, and protein was significantly lower in UNx rats that were treated with JB3 10 microg/kg per day (P: < 0.05) than in saline vehicle controls but was abolished in diabetic rats that were treated with JB3 3 microg/kg per day (P: < 0. 01). Increasing the dose of JB3 to 30 microg/kg per day was associated with a decrease in its inhibitory effect, resulting in bell-shaped dose-response curves. JB3 administration had no effect on the blood glucose concentration or food consumption by either diabetic or nondiabetic animals. The results support the concept of IGF-I as an important mediator of the early renal growth that follows the induction of diabetes or UNx in the rat.


Subject(s)
Diabetes Mellitus, Experimental/physiopathology , Insulin-Like Growth Factor I/pharmacology , Kidney/drug effects , Kidney/growth & development , Nephrectomy , Receptor, IGF Type 1/antagonists & inhibitors , Animals , Cells, Cultured , Diabetes Mellitus/etiology , Diabetes Mellitus/physiopathology , Dose-Response Relationship, Drug , Humans , Insulin-Like Growth Factor I/analogs & derivatives , Male , Opossums , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...