Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Nutr Neurosci ; : 1-19, 2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38576309

ABSTRACT

BACKGROUND: The bed nucleus of the stria terminalis (BNST) is a structure with a peculiar neurochemical composition involved in modulating anxietylike behavior and fear. AIM: The present study investigated the effects on the BNST neurochemical composition and neuronal structure in critical moments of the postnatal period in gestational protein-restricted male rats' offspring. METHODS: Dams were maintained during the pregnancy on isocaloric rodent laboratory chow with standard protein content [NP, 17%] or low protein content [LP, 6%]. BNST from male NP and age-matched LP offspring was studied using the isotropic fractionator method, Neuronal 3D reconstruction, dendritic-tree analysis, blotting analysis, and high-performance liquid chromatography. RESULTS: Serum corticosterone levels were higher in male LP offspring than NP rats in 14-day-old offspring, without any difference in 7-day-old progeny. The BNST total cell number and anterodorsal BNST division volume in LP progeny were significantly reduced on the 14th postnatal day compared with NP offspring. The BNST HPLC analysis from 7 days-old LP revealed increased norepinephrine levels compared to NP progeny. The BNST blot analysis from 7-day-old LP revealed reduced levels of GR and BDNF associated with enhanced CRF1 expression compared to NP offspring. 14-day-old LP offspring showed reduced expression of MR and 5HT1A associated with decreased DOPAC and DOPA turnover levels relative to NP rats. In Conclusion, the BNST cellular and neurochemical changes may represent adaptation during development in response to elevated fetal exposure to maternal corticosteroid levels. In this way, gestational malnutrition alters the BNST content and structure and contributes to already-known behavioral changes.

2.
J Dev Orig Health Dis ; 9(5): 536-543, 2018 10.
Article in English | MEDLINE | ID: mdl-29900811

ABSTRACT

Animal evidence has suggested that maternal emotional and nutritional stress during pregnancy is associated with behavioral outcomes in offspring. The nature of the stresses applied may differ, but it is often assumed that the mother's hippocampus-hypothalamic-pituitary-adrenal (HHPA) axis response releases higher levels of glucocorticoid hormones. The bed nucleus of the stria terminalis (BNST) is in a pivotal position to regulate the HHPA axis and the stress response, and it has been implicated in anxiety behavior. In the current study, to search whether BNST structural changes and neurochemical alterations are associated with anxiety-related behavior in adult gestational protein-restricted offspring relative to an age-matched normal protein diet (NP) rats, we conduct behavioral tests and, BNST dendritic tree analysis by Sholl analysis, associated to immunoblotting-protein quantification [11ß-HSD2, GR, MR, AT1R, 5HT1A and 5HT2A, corticotrophin-releasing factor (CRH) and CRH1]. Dams were maintained either on isocaloric standard rodent chow [with NP content, 17% casein or low protein content (LP), 6% casein] chow throughout their entire pregnancy. Here, in rats subjected to gestational protein restriction, we found: (a) a significant reduction in dendritic length and impoverished dendritic arborization in BNST neurons; (b) an elevated plasmatic corticosterone levels; and (c) associated with enhanced anxiety-like behavior when compared with age-matched NP offspring. Moreover, altered protein (11ß-HSD2, GR, MR and type 1 CRH receptors) expressions may underlie the increase in anxiety-like behavior in LP offspring. This work represents the first demonstration that BNST developmental plasticity by maternal protein restriction, resulting in fine structural changes and neurochemical alterations that are associated with modified behavioral states.


Subject(s)
Anxiety , Diet, Protein-Restricted , Prenatal Exposure Delayed Effects , Septal Nuclei/embryology , Animals , Behavior, Animal , Body Weight , Female , Male , Maternal Nutritional Physiological Phenomena , Nutritional Status , Pregnancy , Rats , Rats, Wistar , Septal Nuclei/pathology
4.
J Dev Orig Health Dis ; 8(1): 89-100, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28051762

ABSTRACT

Emerging evidence highlights the far-reaching consequences of high-fat diet (HFD) and obesity on kidney morphological and functional disorders. In the present study, we aim to evaluate the effects of early HFD intake on renal function and morphology in maternal protein-restricted offspring (LP). LP and normal protein-intake offspring (NP) were fed HFD (LPH and NPH, respectively) or standard rodent (LPN and NPN) diet from the 8th to 13th week of age. Blood pressure, kidney function, immunohistochemistry and scanning electron microscopy were analyzed. Increased total cholesterol and low-density lipoprotein serum levels were observed in LPH offspring. The adiposity index was reduced in the (LPN) group and, conversely, increased in the NPH and LPH groups. Blood pressure was higher beyond the 10th week of age in the LPH group compared with the other groups. Decreased urinary sodium excretion was observed in LP offspring, whereas the HFD-treated groups presented a decreased urine pH in a time-dependent fashion. The LPN, NPH and LPH groups showed increased expression of type 1 angiotensin II (AngII) receptor (AT1R), TGF-ß1, collagen and fibronectin in the kidneys. Moreover, the adult fetal-programmed offspring showed pronounced effacement of the podocyte foot process associated with the rupture of cell membranes and striking urinary protein excretion, exacerbated by HFD treatment. To the best of our knowledge, this is the first study demonstrating that young fetal-programmed offspring submitted to long-term HFD intake have increased susceptibility to renal structural and functional disorders associated with an accentuated stage of fibrosis and tubular dysfunction.


Subject(s)
Diet, High-Fat/adverse effects , Diet, Protein-Restricted/adverse effects , Kidney Diseases/etiology , Kidney Diseases/pathology , Animals , Gestational Age , Male , Mice , Rats, Wistar , Time Factors
5.
Braz J Med Biol Res ; 47(1): 70-9, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24519093

ABSTRACT

Maintenance of thermal homeostasis in rats fed a high-fat diet (HFD) is associated with changes in their thermal balance. The thermodynamic relationship between heat dissipation and energy storage is altered by the ingestion of high-energy diet content. Observation of thermal registers of core temperature behavior, in humans and rodents, permits identification of some characteristics of time series, such as autoreference and stationarity that fit adequately to a stochastic analysis. To identify this change, we used, for the first time, a stochastic autoregressive model, the concepts of which match those associated with physiological systems involved and applied in male HFD rats compared with their appropriate standard food intake age-matched male controls (n=7 per group). By analyzing a recorded temperature time series, we were able to identify when thermal homeostasis would be affected by a new diet. The autoregressive time series model (AR model) was used to predict the occurrence of thermal homeostasis, and this model proved to be very effective in distinguishing such a physiological disorder. Thus, we infer from the results of our study that maximum entropy distribution as a means for stochastic characterization of temperature time series registers may be established as an important and early tool to aid in the diagnosis and prevention of metabolic diseases due to their ability to detect small variations in thermal profile.


Subject(s)
Diet, High-Fat , Dietary Fats/metabolism , Energy Intake/physiology , Energy Metabolism/physiology , Algorithms , Animals , Male , Models, Biological , Rats, Sprague-Dawley , Stochastic Processes , Time Factors
6.
Braz. j. med. biol. res ; 47(1): 70-79, 01/2014. tab, graf
Article in English | LILACS | ID: lil-697675

ABSTRACT

Maintenance of thermal homeostasis in rats fed a high-fat diet (HFD) is associated with changes in their thermal balance. The thermodynamic relationship between heat dissipation and energy storage is altered by the ingestion of high-energy diet content. Observation of thermal registers of core temperature behavior, in humans and rodents, permits identification of some characteristics of time series, such as autoreference and stationarity that fit adequately to a stochastic analysis. To identify this change, we used, for the first time, a stochastic autoregressive model, the concepts of which match those associated with physiological systems involved and applied in male HFD rats compared with their appropriate standard food intake age-matched male controls (n=7 per group). By analyzing a recorded temperature time series, we were able to identify when thermal homeostasis would be affected by a new diet. The autoregressive time series model (AR model) was used to predict the occurrence of thermal homeostasis, and this model proved to be very effective in distinguishing such a physiological disorder. Thus, we infer from the results of our study that maximum entropy distribution as a means for stochastic characterization of temperature time series registers may be established as an important and early tool to aid in the diagnosis and prevention of metabolic diseases due to their ability to detect small variations in thermal profile.


Subject(s)
Animals , Male , Diet, High-Fat , Dietary Fats/metabolism , Energy Intake/physiology , Energy Metabolism/physiology , Algorithms , Models, Biological , Rats, Sprague-Dawley , Stochastic Processes , Time Factors
7.
Int J Dev Neurosci ; 31(3): 151-6, 2013 May.
Article in English | MEDLINE | ID: mdl-23280060

ABSTRACT

Studies have demonstrated that nutrient deficiency during pregnancy or in early postnatal life results in structural abnormalities in the offspring hippocampus and in cognitive impairment. In an attempt to analyze whether gestational protein restriction might induce learning and memory impairments associated with structural changes in the hippocampus, we carried out a detailed morphometric analysis of the hippocampus of male adult rats together with the behavioral characterization of these animals in the Morris water maze (MWM). Our results demonstrate that gestational protein restriction leads to a decrease in total basal dendritic length and in the number of intersections of CA3 pyramidal neurons whereas the cytoarchitecture of CA1 and dentate gyrus remained unchanged. Despite presenting significant structural rearrangements, we did not observe impairments in the MWM test. Considering the clear dissociation between the behavioral profile and the hippocampus neuronal changes, the functional significance of dendritic remodeling in fetal processing remains undisclosed.


Subject(s)
CA3 Region, Hippocampal/pathology , Dendrites/pathology , Diet, Protein-Restricted , Maze Learning/physiology , Neurons/pathology , Prenatal Exposure Delayed Effects/pathology , Animals , Atrophy/pathology , Atrophy/physiopathology , CA3 Region, Hippocampal/physiopathology , Female , Male , Pregnancy , Prenatal Exposure Delayed Effects/physiopathology , Rats , Rats, Wistar
8.
Braz J Med Biol Res ; 43(11): 1010-8, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21049242

ABSTRACT

Maternal dietary protein restriction during pregnancy is associated with low fetal birth weight and leads to renal morphological and physiological changes. Different mechanisms can contribute to this phenotype: exposure to fetal glucocorticoid, alterations in the components of the renin-angiotensin system, apoptosis, and DNA methylation. A low-protein diet during gestation decreases the activity of placental 11ß-hydroxysteroid dehydrogenase, exposing the fetus to glucocorticoids and resetting the hypothalamic-pituitary-adrenal axis in the offspring. The abnormal function/expression of type 1 (AT1(R)) or type 2 (AT2(R)) AngII receptors during any period of life may be the consequence or cause of renal adaptation. AT1(R) is up-regulated, compared with control, on the first day after birth of offspring born to low-protein diet mothers, but this protein appears to be down-regulated by 12 days of age and thereafter. In these offspring, AT2(R) expression differs from control at 1 day of age, but is also down-regulated thereafter, with low nephron numbers at all ages: from the fetal period, at the end of nephron formation, and during adulthood. However, during adulthood, the glomerular filtration rate is not altered, due to glomerulus and podocyte hypertrophy. Kidney tubule transporters are regulated by physiological mechanisms; Na(+)/K(+)-ATPase is inhibited by AngII and, in this model, the down-regulated AngII receptors fail to inhibit Na(+)/K(+)-ATPase, leading to increased Na(+) reabsorption, contributing to the hypertensive status. We also considered the modulation of pro-apoptotic and anti-apoptotic factors during nephrogenesis, since organogenesis depends upon a tight balance between proliferation, differentiation and cell death.


Subject(s)
Hypertension/etiology , Kidney/physiopathology , Pregnancy Complications/physiopathology , Prenatal Exposure Delayed Effects/physiopathology , Protein Deficiency/physiopathology , 11-beta-Hydroxysteroid Dehydrogenases/metabolism , Animals , Animals, Newborn , Apoptosis/physiology , Birth Weight , Diet, Protein-Restricted/adverse effects , Female , Glucocorticoids/metabolism , Humans , Hypertension/physiopathology , Kidney/metabolism , Kidney Glomerulus/metabolism , Kidney Glomerulus/physiopathology , Maternal Nutritional Physiological Phenomena , Pregnancy , Receptors, Angiotensin/metabolism , Renin-Angiotensin System/physiology
9.
Braz. j. med. biol. res ; 43(11): 1010-1018, Nov. 2010. ilus, tab
Article in English | LILACS | ID: lil-564138

ABSTRACT

Maternal dietary protein restriction during pregnancy is associated with low fetal birth weight and leads to renal morphological and physiological changes. Different mechanisms can contribute to this phenotype: exposure to fetal glucocorticoid, alterations in the components of the renin-angiotensin system, apoptosis, and DNA methylation. A low-protein diet during gestation decreases the activity of placental 11ß-hydroxysteroid dehydrogenase, exposing the fetus to glucocorticoids and resetting the hypothalamic-pituitary-adrenal axis in the offspring. The abnormal function/expression of type 1 (AT1R) or type 2 (AT2R) AngII receptors during any period of life may be the consequence or cause of renal adaptation. AT1R is up-regulated, compared with control, on the first day after birth of offspring born to low-protein diet mothers, but this protein appears to be down-regulated by 12 days of age and thereafter. In these offspring, AT2R expression differs from control at 1 day of age, but is also down-regulated thereafter, with low nephron numbers at all ages: from the fetal period, at the end of nephron formation, and during adulthood. However, during adulthood, the glomerular filtration rate is not altered, due to glomerulus and podocyte hypertrophy. Kidney tubule transporters are regulated by physiological mechanisms; Na+/K+-ATPase is inhibited by AngII and, in this model, the down-regulated AngII receptors fail to inhibit Na+/K+-ATPase, leading to increased Na+ reabsorption, contributing to the hypertensive status. We also considered the modulation of pro-apoptotic and anti-apoptotic factors during nephrogenesis, since organogenesis depends upon a tight balance between proliferation, differentiation and cell death.


Subject(s)
Animals , Female , Humans , Pregnancy , Hypertension/etiology , Kidney/physiopathology , Pregnancy Complications/physiopathology , Prenatal Exposure Delayed Effects/physiopathology , Protein Deficiency/physiopathology , Animals, Newborn , /metabolism , Apoptosis/physiology , Birth Weight , Diet, Protein-Restricted/adverse effects , Glucocorticoids/metabolism , Hypertension/physiopathology , Kidney Glomerulus/metabolism , Kidney Glomerulus/physiopathology , Kidney/metabolism , Maternal Nutritional Physiological Phenomena , Receptors, Angiotensin/metabolism , Renin-Angiotensin System/physiology
10.
Braz. j. med. biol. res ; 42(12): 1196-1202, Dec. 2009. ilus, tab
Article in English | LILACS | ID: lil-532292

ABSTRACT

In the present study, we investigated the effects of acute intracerebroventricular (icv) insulin administration on central mechanisms regulating urinary sodium excretion in simultaneously centrally NG-nitro-L-arginine methylester (L-NAME)-injected unanesthetized rats. Male Wistar-Hannover rats were randomly assigned to one of five groups: a) icv 0.15 M NaCl-injected rats (control, N = 10), b) icv dose-response (1.26, 12.6 and 126 ng/3 µL) insulin-injected rats (N = 10), c) rats icv injected with 60 µg L-NAME in combination with NaCl (N = 10) or d) with insulin (N = 10), and e) subcutaneously insulin-injected rats (N = 5). Centrally administered insulin produced an increase in urinary output of sodium (NaCl: 855.6 ± 85.1 Ä percent/min; 126 ng insulin: 2055 ± 310.6 Ä percent/min; P = 0.005) and potassium (NaCl: 460.4 ± 100 Ä percent/min; 126 ng insulin: 669.2 ± 60.8 Ä percent/min; P = 0.025). The urinary sodium excretion response to icv 126 ng insulin microinjection was significantly attenuated by combined administration of L-NAME (126 ng insulin: 1935 ± 258.3 Ä percent/min; L-NAME + 126 ng insulin: 582.3 ± 69.6 Ä percent/min; P = 0.01). Insulin-induced natriuresis occurred by increasing post-proximal sodium excretion, despite an unchanged glomerular filtration rate. Although the rationale for decreased urinary sodium excretion induced by combined icv L-NAME and insulin administration is unknown, it is tempting to suggest that perhaps one of the efferent signals triggered by insulin in the CNS may be nitrergic in nature.


Subject(s)
Animals , Male , Rats , Brain/enzymology , Insulin/pharmacology , Natriuresis/drug effects , Nitric Oxide Synthase/antagonists & inhibitors , Injections, Intraventricular , Insulin/administration & dosage , Microinjections , NG-Nitroarginine Methyl Ester/administration & dosage , Random Allocation , Rats, Wistar
11.
Braz J Med Biol Res ; 42(12): 1196-202, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19893989

ABSTRACT

In the present study, we investigated the effects of acute intracerebroventricular (icv) insulin administration on central mechanisms regulating urinary sodium excretion in simultaneously centrally NG-nitro-L-arginine methylester (L-NAME)-injected unanesthetized rats. Male Wistar-Hannover rats were randomly assigned to one of five groups: a) icv 0.15 M NaCl-injected rats (control, N = 10), b) icv dose-response (1.26, 12.6 and 126 ng/3 microL) insulin-injected rats (N = 10), c) rats icv injected with 60 microg L-NAME in combination with NaCl (N = 10) or d) with insulin (N = 10), and e) subcutaneously insulin-injected rats (N = 5). Centrally administered insulin produced an increase in urinary output of sodium (NaCl: 855.6 +/- 85.1 Delta%/min; 126 ng insulin: 2055 +/- 310.6 Delta%/min; P = 0.005) and potassium (NaCl: 460.4 +/- 100 Delta%/min; 126 ng insulin: 669.2 +/- 60.8 Delta%/min; P = 0.025). The urinary sodium excretion response to icv 126 ng insulin microinjection was significantly attenuated by combined administration of L-NAME (126 ng insulin: 1935 +/- 258.3 Delta%/min; L-NAME + 126 ng insulin: 582.3 +/- 69.6 Delta%/min; P = 0.01). Insulin-induced natriuresis occurred by increasing post-proximal sodium excretion, despite an unchanged glomerular filtration rate. Although the rationale for decreased urinary sodium excretion induced by combined icv L-NAME and insulin administration is unknown, it is tempting to suggest that perhaps one of the efferent signals triggered by insulin in the CNS may be nitrergic in nature.


Subject(s)
Brain/enzymology , Insulin/pharmacology , Natriuresis/drug effects , Nitric Oxide Synthase/antagonists & inhibitors , Animals , Injections, Intraventricular , Insulin/administration & dosage , Male , Microinjections , NG-Nitroarginine Methyl Ester/administration & dosage , Random Allocation , Rats , Rats, Wistar
12.
Transplant Proc ; 40(10): 3311-5, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19100379

ABSTRACT

BACKGROUND: Proximal tubule cells have specialized apical membranes with microvilli that provide an extensive surface area for unidirectional transport of solute from lumen to blood. The major structural solute component is F-actin, which interacts with transmembrane proteins, including ion transport molecules related to normal absorptive and secretory functions. Our study was to evaluate F-actin and fluid absorption (Jv) in proximal tubules after exposure to preservation solutions. METHODS: In vitro microperfusion technique and immunohistochemistry analysis. RESULTS: 1. Absorptions were similar in 1- and 24-hour-preserved tubules, as well as in fresh tubules. The exception was tubules for 24 hours in Euro-Collins solution, which did not show absorption, suggesting that it was affected. 2. Fluorescence intensity of actin tubules preserved for 1 hour in both solutions showed similar values to each other and to the control group; tubules preserved for 24 hours in both solutions were similar to each other, although statistically different than the control group and those preserved for 1 hour in Belzer (UW) solution. CONCLUSION: There were differences among groups in the distribution of F-actin; Jv values were different for 24-hour preservation in each solution, whereas fluorescence intensity was similar in both 24-hour solutions. Thus, actin cytoskeleton was not responsible for it, because 24-hour preservation in UW showed Jv results comparable to the control group.


Subject(s)
Actins/physiology , Cytoskeleton/physiology , Kidney Tubules, Proximal/physiology , Organ Preservation Solutions/pharmacology , Organ Preservation/methods , Absorption , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/ultrastructure , Actins/ultrastructure , Adenosine/pharmacology , Allopurinol/pharmacology , Animals , Cytoskeleton/drug effects , Cytoskeleton/ultrastructure , Glutathione/pharmacology , Insulin/pharmacology , Kidney Tubules, Collecting/drug effects , Kidney Tubules, Collecting/physiology , Kidney Tubules, Collecting/ultrastructure , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/ultrastructure , Male , Microscopy, Confocal/methods , Microvilli/drug effects , Microvilli/ultrastructure , Models, Animal , Organ Preservation Solutions/pharmacokinetics , Perfusion/methods , Rabbits , Raffinose/pharmacology
13.
Braz. j. med. biol. res ; 38(12): 1817-1824, Dec. 2005. ilus
Article in English | LILACS | ID: lil-417190

ABSTRACT

The present study evaluated the acute effect of the intraperitoneal (ip) administration of a whey protein hydrolysate (WPH) on systolic arterial blood pressure (SBP) and renal sodium handling by conscious spontaneously hypertensive rats (SHR). The ip administration of WPH in a volume of 1 ml dose-dependently lowered the SBP in SHR 2 h after administration at doses of 0.5 g/kg (0.15 M NaCl: 188.5 ± 9.3 mmHg vs WPH: 176.6 ± 4.9 mmHg, N = 8, P = 0.001) and 1.0 g/kg (0.15 M NaCl: 188.5 ± 9.3 mmHg vs WPH: 163.8 ± 5.9 mmHg, N = 8, P = 0.0018). Creatinine clearance decreased significantly (P = 0.0084) in the WPH-treated group (326 ± 67 æL min-1 100 g body weight-1) compared to 0.15 M NaCl-treated (890 ± 26 æL min-1 100 g body weight-1) and captopril-treated (903 ± 72 æL min-1 100 g body weight-1) rats. The ip administration of 1.0 g WPH/kg also decreased fractional sodium excretion to 0.021 ± 0.019 percent compared to 0.126 ± 0.041 and 0.66 ± 0.015 percent in 0.15 M NaCl and captopril-treated rats, respectively (P = 0.033). Similarly, the fractional potassium excretion in WPH-treated rats (0.25 ± 0.05 percent) was significantly lower (P = 0.0063) than in control (0.91 ± 0.15 percent) and captopril-treated rats (1.24 ± 0.30 percent), respectively. The present study shows a decreased SBP in SHR after the administration of WPH associated with a rise in tubule sodium reabsorption despite an angiotensin I-converting enzyme (ACE)-inhibiting in vitro activity (IC50 = 0.68 mg/mL). The present findings suggest a pathway involving ACE inhibition but measurements of plasma ACE activity and angiotensin II levels are needed to support this suggestion.


Subject(s)
Animals , Male , Rats , Protein Hydrolysates/pharmacology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Arterial Pressure/drug effects , Milk Proteins/pharmacology , Captopril/pharmacology , Electrophoresis, Capillary , Protein Hydrolysates/administration & dosage , Kidney Function Tests , Potassium/urine , Milk Proteins/administration & dosage , Rats, Inbred SHR , Sodium/urine
14.
Braz J Med Biol Res ; 38(12): 1817-24, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16302096

ABSTRACT

The present study evaluated the acute effect of the intraperitoneal (ip) administration of a whey protein hydrolysate (WPH) on systolic arterial blood pressure (SBP) and renal sodium handling by conscious spontaneously hypertensive rats (SHR). The ip administration of WPH in a volume of 1 ml dose-dependently lowered the SBP in SHR 2 h after administration at doses of 0.5 g/kg (0.15 M NaCl: 188.5 +/- 9.3 mmHg vs WPH: 176.6 +/- 4.9 mmHg, N = 8, P = 0.001) and 1.0 g/kg (0.15 M NaCl: 188.5 +/- 9.3 mmHg vs WPH: 163.8 +/- 5.9 mmHg, N = 8, P = 0.0018). Creatinine clearance decreased significantly (P = 0.0084) in the WPH-treated group (326 +/- 67 microL min-1 100 g body weight-1) compared to 0.15 M NaCl-treated (890 +/- 26 microL min-1 100 g body weight-1) and captopril-treated (903 +/- 72 microL min-1 100 g body weight-1) rats. The ip administration of 1.0 g WPH/kg also decreased fractional sodium excretion to 0.021 +/- 0.019% compared to 0.126 +/- 0.041 and 0.66 +/- 0.015% in 0.15 M NaCl and captopril-treated rats, respectively (P = 0.033). Similarly, the fractional potassium excretion in WPH-treated rats (0.25 +/- 0.05%) was significantly lower (P = 0.0063) than in control (0.91 +/- 0.15%) and captopril-treated rats (1.24 +/- 0.30%), respectively. The present study shows a decreased SBP in SHR after the administration of WPH associated with a rise in tubule sodium reabsorption despite an angiotensin I-converting enzyme (ACE)-inhibiting in vitro activity (IC50 = 0.68 mg/mL). The present findings suggest a pathway involving ACE inhibition but measurements of plasma ACE activity and angiotensin II levels are needed to support this suggestion.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Blood Pressure/drug effects , Milk Proteins/pharmacology , Protein Hydrolysates/pharmacology , Animals , Captopril/pharmacology , Electrophoresis, Capillary , Kidney Function Tests , Male , Milk Proteins/administration & dosage , Potassium/urine , Protein Hydrolysates/administration & dosage , Rats , Rats, Inbred SHR , Sodium/urine , Whey Proteins
15.
Diabetologia ; 46(12): 1629-40, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14600817

ABSTRACT

AIM/HYPOTHESIS: By acting in the brain, insulin suppresses food intake. However, little is known with regard to insulin signalling in the hypothalamus in insulin-resistant states. METHODS: Western blotting, immunohistochemistry and polymerase chain reaction assays were combined to compare in vivo hypothalamic insulin signalling through the PI3-kinase and MAP kinase pathways between lean and obese Zucker rats. RESULTS: Intracerebroventricular insulin infusion reduced food intake in lean rats to a greater extent than that observed in obese rats, and pre-treatment with PI3-kinase inhibitors prevented insulin-induced anorexia. The relative abundance of IRS-2 was considerably higher than that of IRS-1 in hypothalamus of both lean and obese rats. Insulin-stimulated phosphorylation of IR, IRS-1/2, the associations of PI 3-kinase to IRS-1/2 and phosphorylation of Akt in hypothalamus were decreased in obese rats compared to lean rats. These effects seem to be mediated by increased phosphoserine content of IR, IRS-1/2 and decreased protein levels of IRS-1/2 in obese rats. In contrast, insulin stimulated the phosphorylation of MAP kinase equally in lean and obese rats. CONCLUSION/INTERPRETATION: This study provides direct measurements of insulin signalling in hypothalamus, and documents selective resistance to insulin signalling in hypothalamus of Zucker rats. These findings provide support for the hypothesis that insulin could have anti-obesity actions mediated by the PI3-kinase pathway, and that impaired insulin signalling in hypothalamus could play a role in the development of obesity in this animal model of insulin-resistance.


Subject(s)
Hypothalamus/physiopathology , Insulin/pharmacology , Obesity/physiopathology , Signal Transduction/physiology , Animals , Blood Glucose/metabolism , Body Weight , Injections, Intraventricular , Insulin/administration & dosage , Insulin Receptor Substrate Proteins , Intracellular Signaling Peptides and Proteins , Male , Obesity/genetics , Phosphoproteins/metabolism , Phosphorylation , Phosphoserine/metabolism , Rats , Rats, Zucker , Reference Values
16.
Braz. j. med. biol. res ; 36(9): 1193-1199, Sept. 2003. ilus, tab
Article in English | LILACS | ID: lil-342845

ABSTRACT

Recent evidence suggests that insulin may influence many brain functions. It is known that intracerebroventricular (icv) injection of nondiabetogenic doses of streptozotocin (STZ) can damage insulin receptor signal transduction. In the present study, we examined the functional damage to the brain insulin receptors on central mechanisms regulating glomerular filtration rate and urinary sodium excretion, over four periods of 30 min, in response to 3 æl insulin or 0.15 NaCl (vehicle) injected icv in STZ-treated freely moving Wistar-Hannover rats (250-300 g). The icv cannula site was visually confirmed by 2 percent Evans blue infusion. Centrally administered insulin (42.0 ng/æl) increased the urinary output of sodium (from 855.6 ± 85.1 to 2055 ± 310.6 delta percent/min; N = 11) and potassium (from 460.4 ± 100 to 669 ± 60.8 delta percent/min; N = 11). The urinary sodium excretion response to icv insulin microinjection was markedly attenuated by previous central STZ (100 æg/3 æl) administration (from 628 ± 45.8 to 617 ± 87.6 delta percent/min; N = 5) or by icv injection of a dopamine antagonist, haloperidol (4 æg/3 æl) (from 498 ± 39.4 to 517 ± 73.2 delta percent/min; N = 5). Additionally, insulin-induced natriuresis occurred by increased post-proximal tubule sodium rejection, despite an unchanged glomerular filtration rate. Excluding the possibility of a direct action of STZ on central insulin receptor-carrying neurons, the current data suggest that the insulin-sensitive response may be processed through dopaminergic D1 receptors containing neuronal pathways


Subject(s)
Animals , Male , Rats , Brain , Glomerular Filtration Rate , Insulin , Natriuresis , Receptor, Insulin , Signal Transduction , Antibiotics, Antineoplastic , Injections, Intraventricular , Injections, Subcutaneous , Rats, Wistar , Streptozocin , Time Factors
17.
Braz J Med Biol Res ; 36(9): 1193-9, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12937785

ABSTRACT

Recent evidence suggests that insulin may influence many brain functions. It is known that intracerebroventricular (icv) injection of nondiabetogenic doses of streptozotocin (STZ) can damage insulin receptor signal transduction. In the present study, we examined the functional damage to the brain insulin receptors on central mechanisms regulating glomerular filtration rate and urinary sodium excretion, over four periods of 30 min, in response to 3 microl insulin or 0.15 NaCl (vehicle) injected icv in STZ-treated freely moving Wistar-Hannover rats (250-300 g). The icv cannula site was visually confirmed by 2% Evans blue infusion. Centrally administered insulin (42.0 ng/ micro l) increased the urinary output of sodium (from 855.6 85.1 to 2055 310.6 delta%/min; N = 11) and potassium (from 460.4 100 to 669 60.8 delta%/min; N = 11). The urinary sodium excretion response to icv insulin microinjection was markedly attenuated by previous central STZ (100 micro g/3 micro l) administration (from 628 45.8 to 617 87.6 delta%/min; N = 5) or by icv injection of a dopamine antagonist, haloperidol (4 micro g/3 micro l) (from 498 +/- 39.4 to 517 +/- 73.2 delta%/min; N = 5). Additionally, insulin-induced natriuresis occurred by increased post-proximal tubule sodium rejection, despite an unchanged glomerular filtration rate. Excluding the possibility of a direct action of STZ on central insulin receptor-carrying neurons, the current data suggest that the insulin-sensitive response may be processed through dopaminergic D1 receptors containing neuronal pathways.


Subject(s)
Brain/drug effects , Glomerular Filtration Rate/drug effects , Insulin/administration & dosage , Natriuresis/drug effects , Receptor, Insulin/drug effects , Signal Transduction/drug effects , Animals , Antibiotics, Antineoplastic , Injections, Intraventricular , Injections, Subcutaneous , Male , Rats , Rats, Wistar , Streptozocin , Time Factors
18.
Phytomedicine ; 10(2-3): 127-31, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12725565

ABSTRACT

This study evaluated the effect of oral crude Vernonia polyanthes Less. hydroalcoholic extract administration (CHE, 0.5 and 1.0 g/kg body wt., daily for 7 days) on arterial blood pressure and renal sodium excretion in conscious rats. CHE administration decreased arterial blood pressure dose-dependently followed by a significant rise in creatinine clearance and a fall in fractional post-proximal sodium excretion was compared to the control group. These results suggest that blood pressure decrease induced by the oral crude Vernonia hydroalcoholic extract may be blunted by reduction of the post-proximal renal sodium excretion. Thus, the present study shows that Vernonia extract is a potential vasodilatation agent in normotensive rats without any effects on renal tubule autoregulation mechanisms.


Subject(s)
Blood Pressure/drug effects , Plant Extracts/pharmacology , Sodium/metabolism , Vernonia , Animals , Creatinine/metabolism , Male , Plant Extracts/administration & dosage , Plant Leaves/chemistry , Rats , Rats, Wistar , Sodium/urine
19.
Braz. j. med. biol. res ; 34(9): 1155-1160, Sept. 2001. tab
Article in English | LILACS | ID: lil-290401

ABSTRACT

Rats fed a high-fructose diet represent an animal model for insulin resistance and hypertension. We recently showed that a high-fructose diet containing vegetable oil but a normal sodium/potassium ratio induced mild insulin resistance with decreased insulin receptor substrate-1 tyrosine phosphorylation in the liver and muscle of normal rats. In the present study, we examined the mean blood pressure, serum lipid levels and insulin sensitivity by estimating in vivo insulin activity using the 15-min intravenous insulin tolerance test (ITT, 0.5 ml of 6 æg insulin, iv) followed by calculation of the rate constant for plasma glucose disappearance (Kitt) in male Wistar-Hannover rats (110-130 g) randomly divided into four diet groups: control, 1:3 sodium/potassium ratio (R Na:K) diet (C 1:3 R Na:K); control, 1:1 sodium/potassium ratio diet (CNa 1:1 R Na:K); high-fructose, 1:3 sodium/potassium ratio diet (F 1:3 R Na:K), and high-fructose, 1:1 sodium/potassium ratio diet (FNa 1:1 R Na:K) for 28 days. The change in R Na:K for the control and high-fructose diets had no effect on insulin sensitivity measured by ITT. In contrast, the 1:1 R Na:K increased blood pressure in rats receiving the control and high-fructose diets from 117 + or - 3 and 118 + or - 3 mmHg to 141 + or - 4 and 132 + or - 4 mmHg (P<0.05), respectively. Triacylglycerol levels were higher in both groups treated with a high-fructose diet when compared to controls (C 1:3 R Na:K: 1.2 + or - 0.1 mmol/l vs F 1:3 R Na:K: 2.3 + or - 0.4 mmol/l and CNa 1:1 R Na:K: 1.2 + or - 0.2 mmol/l vs FNa 1:1 R Na:K: 2.6 + or - 0.4 mmol/l, P<0.05). These data suggest that fructose alone does not induce hyperinsulinemia or hypertension in rats fed a normal R Na:K diet, whereas an elevation of sodium in the diet may contribute to the elevated blood pressure in this animal model


Subject(s)
Animals , Male , Rats , Blood Pressure , Diet , Fructose/physiology , Insulin Resistance , Blood Glucose/analysis , Hyperinsulinism/etiology , Hypertension/etiology , Hypertriglyceridemia/etiology , Lipids/blood , Potassium/administration & dosage , Rats, Wistar , Sodium/administration & dosage
20.
Braz. j. med. biol. res ; 33(3): 347-54, Mar. 2000. tab, graf
Article in English | LILACS | ID: lil-255054

ABSTRACT

The role of sympathetic nerve activity in the changes in arterial blood pressure and renal function caused by the chronic administration of NG-nitro-L-arginine methyl ester (L-NAME), an inhibitor of nitric oxide (NO) synthesis, was examined in sham and bilaterally renal denervated rats. Several studies have demonstrated that sympathetic nerve activity is elevated acutely after L-NAME administration. To evaluate the role of renal nerve activity in L-NAME-induced hypertension, we compared the blood pressure response in four groups (N = 10 each) of male Wistar-Hannover rats weighing 200 to 250 g: 1) sham-operated vehicle-treated, 2) sham-operated L-NAME-treated, 3) denervated vehicle-treated, and 4) denervated L-NAME-treated rats. After renal denervation or sham surgery, one control week was followed by three weeks of oral administration of L-NAME by gavage. Arterial pressure was measured weekly in conscious rats by a tail-cuff method and renal function tests were performed in individual metabolic cages 0, 7, 14 and 21 days after the beginning of L-NAME administration. L-NAME (60 mg kg-1 day-1) progressively increased arterial pressure from 108 + or - 6.0 to 149 + or - 12 mmHg (P<0.05) in the sham-operated group by the third week of treatment which was accompanied by a fall in creatinine clearance from 336 + or - 18 to 222 + or - 59 µl min-1 100 g body weight-1 (P<0.05) and a rise in fractional urinary sodium excretion from 0.2 + or - 0.04 to 1.62 + or - 0.35 per cent (P<0.05) and in sodium post-proximal fractional excretion from 0.54 + or - 0.09 to 4.7 + or - 0.86 per cent (P<0.05). The development of hypertension was significantly delayed and attenuated in denervated L-NAME-treated rats. This was accompanied by a striking additional increase in fractional renal sodium and potassium excretion from 0.2 + or - 0.04 to 4.5 + or - 1.6 per cent and from 0.1 + or - 0.015 to 1.21 + or - 0.37 per cent, respectively, and an enhanced post-proximal sodium excretion compared to the sham-operated group. These differences occurred despite an unchanged creatinine clearance and Na+ filtered load. These results suggest that bilateral renal denervation delayed and attenuated the L-NAME-induced hypertension by promoting an additional decrease in tubule sodium reabsorption in the post-proximal segments of nephrons. Much of the hypertension caused by chronic NO synthesis inhibition is thus dependent on renal nerve activity.


Subject(s)
Animals , Male , Rats , Blood Pressure/drug effects , Denervation , Enzyme Inhibitors/pharmacology , Kidney/innervation , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/antagonists & inhibitors , Sodium/metabolism , Enzyme Inhibitors/adverse effects , Hypertension/chemically induced , Kidney/chemistry , Kidney/physiology , NG-Nitroarginine Methyl Ester/adverse effects , Nitric Oxide/biosynthesis , Rats, Wistar , Sodium/urine , Sympathetic Nervous System/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...