Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters










Publication year range
1.
Antioxidants (Basel) ; 11(9)2022 Sep 13.
Article in English | MEDLINE | ID: mdl-36139877

ABSTRACT

Carob, the fruit of Ceratonia siliqua L. exerts antidiabetic, anti-inflammatory, and antioxidant effects and could be a useful strategy for the treatment and/or prevention of metabolic syndrome (MetS). The aim of this study was to analyze whether supplementation with a carob fruit extract (CSAT+®), alone or in combination with aerobic training, accelerates the recovery of cardiometabolic health in mice with MetS subjected to a caloric restriction. For this purpose, mice were fed with a high fat (58% kcal from fat)/high sugar diet for 23 weeks to induce MetS. During the next two weeks, mice with MetS were switched to a diet with a lower caloric content (25% kcal from fat) supplemented or not with CSAT+® (4.8%) and/or subjected to aerobic training. Both caloric reduction and aerobic training improved the lipid profile and attenuated MetS-induced insulin resistance measured as HOMA-IR. However, only supplementation with CSAT+® enhanced body weight loss, increased the circulating levels of adiponectin, and lowered the plasma levels of IL-6. Moreover, CSAT+® supplementation was the only effective strategy to reduce the weight of epidydimal adipose tissue and to improve insulin sensitivity in the liver and in skeletal muscle. Although all interventions improved endothelial function in aorta segments, only supplementation with CSAT+® reduced obesity-induced hypertension, prevented endothelial dysfunction in mesenteric arteries, and decreased the vascular response of aorta segments to the vasoconstrictor AngII. The beneficial cardiometabolic effects of CSAT+® supplementation, alone or in combination with aerobic training, were associated with decreased mRNA levels of pro-inflammatory markers such as MCP-1, TNFα, IL-1ß, and IL-6 and with increased gene expression of antioxidant enzymes, such as GSR, GPX-3, and SOD-1 in the liver, gastrocnemius, retroperitoneal adipose tissue, and aorta. In conclusion, supplementation with CSAT+®, alone or in combination with aerobic training, to mice with MetS subjected to caloric restriction for two weeks enhances body weight loss, improves the lipid profile and insulin sensitivity, and exerts antihypertensive effects through its anti-inflammatory and antioxidant properties.

2.
Antioxidants (Basel) ; 11(4)2022 Mar 30.
Article in English | MEDLINE | ID: mdl-35453365

ABSTRACT

In recent years, gut dysbiosis has been related to some peripheral vascular alterations linked to hypertension. In this work, we explore whether gut dysbiosis is related to vascular innervation dysfunction and altered nitric oxide (NO) production in the superior mesenteric artery, one of the main vascular beds involved in peripheral vascular resistance. For this purpose, we used spontaneously hypertensive rats, either treated or not with the commercial synbiotic formulation Prodefen® (108 colony forming units/day, 4 weeks). Prodefen® diminished systolic blood pressure and serum endotoxin, as well as the vasoconstriction elicited by electrical field stimulation (EFS), and enhanced acetic and butyric acid in fecal samples, and the vasodilation induced by the exogenous NO donor DEA-NO. Unspecific nitric oxide synthase (NOS) inhibitor L-NAME increased EFS-induced vasoconstriction more markedly in rats supplemented with Prodefen®. Both neuronal NO release and neuronal NOS activity were enhanced by Prodefen®, through a hyperactivation of protein kinase (PK)A, PKC and phosphatidylinositol 3 kinase-AKT signaling pathways. The superoxide anion scavenger tempol increased both NO release and DEA-NO vasodilation only in control animals. Prodefen® caused an increase in both nuclear erythroid related factor 2 and superoxide dismutase activities, consequently reducing both superoxide anion and peroxynitrite releases. In summary, Prodefen® could be an interesting non-pharmacological approach to ameliorate hypertension.

4.
Int J Mol Sci ; 22(2)2021 Jan 08.
Article in English | MEDLINE | ID: mdl-33430047

ABSTRACT

We aimed to determine whether an experimental model of hyperthyroidism could alter the function of sympathetic and nitrergic components of mesenteric innervation. For this purpose, male Wistar rats were divided into (1) control rats (CT) and (2) rats infused with L-Thyroxine (HT). Body weight gain and adipose tissue accumulation were lower in HT rats, while systolic blood pressure and citrate synthase activity in the soleus muscle were increased by HT. In segments from the superior mesenteric artery, the application of an electrical field stimulation (EFS) induced a vasoconstrictor response, which was lower in arteries from HT animals. The alpha-adrenoceptor antagonist phentolamine diminished EFS-induced vasoconstriction to a lower extent in HT arteries, while the purinergic receptor antagonist suramin reduced contractile response to EFS only in segments from CT. In line with this, noradrenaline release, tyrosine hydroxylase expression and activation and dopamine ß hydroxylase expression were diminished in HT. The unspecific nitric oxide synthase (NOS) inhibitor L-NAME increased EFS-induced vasoconstriction more markedly in segments from HT rats. NO release was enhanced in HT, probably due to an enhancement in neuronal NOS activity, in which a hyperactivation of both PKC and PI3K-AKT signaling pathways might play a relevant role. In conclusion, perivascular mesenteric innervation might contribute to reduce the vascular resistance observed in hyperthyroidism.


Subject(s)
Body Weight/drug effects , Hyperthyroidism/genetics , Nitric Oxide Synthase/genetics , Nitric Oxide/genetics , Adipose Tissue/drug effects , Adipose Tissue/growth & development , Animals , Body Weight/genetics , Disease Models, Animal , Electric Stimulation , Humans , Hyperthyroidism/metabolism , Hyperthyroidism/pathology , Mesenteric Arteries/drug effects , Mesenteric Arteries/growth & development , Mesenteric Veins/drug effects , Mesenteric Veins/growth & development , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Rats , Rats, Wistar , Thyroxine/pharmacology , Vasoconstriction/genetics
5.
Am J Physiol Heart Circ Physiol ; 320(2): H563-H574, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33164582

ABSTRACT

Heart failure (HF) is associated with neurohumoral activation, which in turn leads to an increased peripheral resistance. In mesenteric vasculature, perivascular innervation plays relevant role maintaining vascular tonus and resistance. Therefore, we aimed to determine the possible alterations in superior mesenteric artery (SMA) perivascular innervation function in HF rats. HF was induced by coronary artery occlusion in male Wistar rats, and sham-operated (SO) rats were used as controls. After 12 wk, a greater vasoconstrictor response to electrical field stimulation (EFS) was observed in endothelium-intact and endothelium-denuded SMA of HF rats. Alpha-adrenoceptor antagonist phentolamine diminished this response in a higher magnitude in HF than in SO animals. However, the noradrenaline (NA) reuptake inhibitor desipramine increased EFS-induced vasoconstriction more in segments from HF rats. Besides, EFS-induced NA release was greater in HF animals, due to a higher tyrosine hydroxylase expression and activity. P2 purinoceptor antagonist suramin reduced EFS-induced vasoconstriction only in segments from SO rats, and adenosine 5'-triphosphate (ATP) release was lower in HF than in SO. Moreover, nitric oxide (NO) synthase inhibitor Nω-nitro-L-arginine methyl ester (L-NAME) enhanced EFS-induced vasoconstriction in a similar extent in both groups. HF was not associated with changes in EFS-induced NO release or the vasodilator response to NO donor sodium nitroprusside. In conclusion, HF postmyocardial infarction enhanced noradrenergic function and diminished purinergic cotransmission in SMA and did not change nitrergic innervation. The net effect was an increased sympathetic participation on the EFS-induced vasoconstriction that could help to understand the neurotransduction involved on the control of vascular tonus in HF.NEW & NOTEWORTHY This study reinforces the pivotal role of noradrenergic innervation in the regulation of mesenteric vascular tone in a rat model of heart failure. Moreover, our results highlight the counteracting role of ATP and NA reuptake, and help to understand the signaling pathways involved on the control of vascular tonus and resistance in heart failure postmyocardial infarction.


Subject(s)
Adenosine Triphosphate/metabolism , Heart Failure/metabolism , Norepinephrine/metabolism , Synaptic Transmission , Adrenergic Uptake Inhibitors/pharmacology , Adrenergic alpha-Antagonists/pharmacology , Animals , Desipramine/pharmacology , Enzyme Inhibitors/pharmacology , Heart Failure/physiopathology , Male , Mesenteric Arteries/drug effects , Mesenteric Arteries/metabolism , Mesenteric Arteries/physiopathology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Donors/pharmacology , Nitroprusside/pharmacology , Phentolamine/pharmacology , Purinergic P2 Receptor Antagonists/pharmacology , Rats , Rats, Wistar , Suramin/pharmacology , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/metabolism , Sympathetic Nervous System/physiopathology , Vasoconstriction
6.
Front Physiol ; 11: 593371, 2020.
Article in English | MEDLINE | ID: mdl-33329042

ABSTRACT

The acute-on-chronic liver failure (ACLF) is a syndrome characterized by liver decompensation, hepatic encephalopathy (HE) and high mortality. We aimed to determine the mechanisms implicated in the development of HE-associated cerebral vasculopathy in a microsurgical liver cholestasis (MHC) model of ACLF. Microsurgical liver cholestasis was induced by ligating and extracting the common bile duct and four bile ducts. Sham-operated and MHC rats were maintained for eight postoperative weeks Bradykinin-induced vasodilation was greater in middle cerebral arteries from MHC rats. Both Nω-Nitro-L-arginine methyl ester and indomethacin diminished bradykinin-induced vasodilation largely in arteries from MHC rats. Nitrite and prostaglandin (PG) F1α releases were increased, whereas thromboxane (TX) B2 was not modified in arteries from MHC. Expressions of endothelial nitric oxide synthase (eNOS), inducible NOS, and cyclooxygenase (COX) 2 were augmented, and neuronal NOS (nNOS), COX-1, PGI2 synthase, and TXA2S were unmodified. Phosphorylation was augmented for eNOS and unmodified for nNOS. Altogether, these endothelial alterations might collaborate to increase brain blood flow in HE.

7.
Curr Pharm Des ; 26(30): 3633-3651, 2020.
Article in English | MEDLINE | ID: mdl-32242780

ABSTRACT

Cardiovascular diseases (CVD) are considered a major health problem worldwide, being the main cause of mortality in developing and developed countries. Endothelial dysfunction, characterized by a decline in nitric oxide production and/or bioavailability, increased oxidative stress, decreased prostacyclin levels, and a reduction of endothelium-derived hyperpolarizing factor is considered an important prognostic indicator of various CVD. Changes in cyclic nucleotides production and/ or signalling, such as guanosine 3', 5'-monophosphate (cGMP) and adenosine 3', 5'-monophosphate (cAMP), also accompany many vascular disorders that course with altered endothelial function. Phosphodiesterases (PDE) are metallophosphohydrolases that catalyse cAMP and cGMP hydrolysis, thereby terminating the cyclic nucleotide-dependent signalling. The development of drugs that selectively block the activity of specific PDE families remains of great interest to the research, clinical and pharmaceutical industries. In the present review, we will discuss the effects of PDE inhibitors on CVD related to altered endothelial function, such as atherosclerosis, diabetes mellitus, arterial hypertension, stroke, aging and cirrhosis. Multiple evidences suggest that PDEs inhibition represents an attractive medical approach for the treatment of endothelial dysfunction-related diseases. Selective PDE inhibitors, especially PDE3 and PDE5 inhibitors are proposed to increase vascular NO levels by increasing antioxidant status or endothelial nitric oxide synthase expression and activation and to improve the morphological architecture of the endothelial surface. Thereby, selective PDE inhibitors can improve the endothelial function in various CVD, increasing the evidence that these drugs are potential treatment strategies for vascular dysfunction and reinforcing their potential role as an adjuvant in the pharmacotherapy of CVD.


Subject(s)
Cyclic GMP , Phosphoric Diester Hydrolases , Biological Factors , Humans , Nitric Oxide , Nucleotides, Cyclic
8.
Nutrients ; 12(1)2020 Jan 01.
Article in English | MEDLINE | ID: mdl-31906276

ABSTRACT

A high fat diet (HFD) intake is crucial for the development and progression of metabolic syndrome (MtS). Increasing evidence links gut dysbiosis with the metabolic and vascular alterations associated with MtS. Here we studied the use of a combination of various probiotic strains together with a prebiotic (synbiotic) in a commercially available Prodefen® Plus. MtS was induced by HFD (45%) in male Wistar rats. Half of the MtS animals received Prodefen® Plus for 4 weeks. At 12 weeks, we observed an increase in body weight, together with the presence of insulin resistance, liver steatosis, hypertriglyceridemia and hypertension in MtS rats. Prodefen® Plus supplementation did not affect the body weight gain but ameliorated all the MtS-related symptoms. Moreover, the hypertension induced by HFD is caused by a diminished both nitric oxide (NO) functional role and release probably due to a diminished neuronal nitric oxide synthase (nNOS) activation by protein kinase A (PKA) pathway. Prodefen® Plus supplementation for 4 weeks recovered the NO function and release and the systolic blood pressure was returned to normotensive values as a result. Overall, supplementation with Prodefen® Plus could be considered an interesting non-pharmacological approach in MtS.


Subject(s)
Blood Vessels/physiopathology , Cyclic AMP-Dependent Protein Kinases/physiology , Metabolic Syndrome/enzymology , Nitric Oxide Synthase/physiology , Synbiotics/administration & dosage , Animals , Blood Pressure/physiology , Blood Vessels/enzymology , Body Weight , Diet, High-Fat , Dietary Supplements , Disease Models, Animal , Hypertension/enzymology , Hypertension/etiology , Hypertension/physiopathology , Insulin Resistance , Male , Metabolic Syndrome/etiology , Metabolic Syndrome/physiopathology , Probiotics/administration & dosage , Rats , Rats, Wistar
10.
Arch Med Res ; 50(5): 271-284, 2019 07.
Article in English | MEDLINE | ID: mdl-31593852

ABSTRACT

Chronic inflammatory liver disease with an acute deterioration of liver function is named acute-on-chronic inflammation and could be regulated by the metabolic impairments related to the liver dysfunction. In this way, the experimental cholestasis model is excellent for studying metabolism in both types of inflammatory responses. Along the evolution of this model, the rats develop biliary fibrosis and an acute-on-chronic decompensation. The acute decompensation of the liver disease is associated with encephalopathy, ascites, acute renal failure, an acute phase response and a splanchnic increase of pro- and anti-inflammatory cytokines. This multiorgan inflammatory dysfunction is mainly associated with a splanchnic and systemic metabolic switch with dedifferentiation of the epithelial, endothelial and mesothelial splanchnic barriers. Furthermore, a splanchnic infiltration by mast cells occurs, which suggests that these cells could carry out a compensatory metabolic role, especially through the modulation of hepatic and extrahepatic mitochondrial-peroxisome crosstalk. For this reason, we propose the hypothesis that mastocytosis in the acute-on-chronic hepatic insufficiency could represent the development of a survival metabolic mechanisms that mitigates the noxious effect of the hepatic functional deficit. A better understanding the pathophysiological response of the mast cells in liver insufficiency and portal hypertension would help to find new pathways for decreasing the high morbidity and mortality rate of these patients.


Subject(s)
Acute-On-Chronic Liver Failure/metabolism , Acute-On-Chronic Liver Failure/therapy , Inflammation/metabolism , Animals , Humans , Rats
11.
Cells ; 8(7)2019 06 29.
Article in English | MEDLINE | ID: mdl-31261968

ABSTRACT

Portal hypertension is a common complication of liver disease, either acute or chronic. Consequently, in chronic liver disease, such as the hypertensive mesenteric venous pathology, the coexisting inflammatory response is classically characterized by the splanchnic blood circulation. However, a vascular lymphatic pathology is produced simultaneously with the splanchnic arterio-venous impairments. The pathological increase of the mesenteric venous pressure, by mechanotransduction of the venous endothelium hyperpressure, causes an inflammatory response involving the subendothelial mast cells and the lymphatic endothelium of the intestinal villi lacteal. In portal hypertension, the intestinal lymphatic inflammatory response through the development of mesenteric-systemic lymphatic collateral vessels favors the systemic diffusion of substances with a molecular pattern associated with damage and pathogens of intestinal origin. When the chronic hepatic insufficiency worsens the portal hypertensive inflammatory response, the splanchnic lymphatic system transports the hyperplasied intestinal mast cells to the mesenteric lymphatic complex. Then, an acquired immune response regulating a new hepato-intestinal metabolic scenario is activated. Therefore, reduction of the hepatic metabolism would reduce its key centralized functions, such as the metabolic, detoxifying and antioxidant functions which would try to be substituted by their peroxisome activity, among other functions of the mast cells.


Subject(s)
Hypertension, Portal/immunology , Inflammation/immunology , Lymphatic Vessels/cytology , Mast Cells/immunology , Splanchnic Circulation/immunology , Humans , Hypertension, Portal/pathology , Inflammation/pathology , Intestinal Mucosa/immunology , Lymphatic Vessels/immunology , Lymphatic Vessels/pathology , Mechanotransduction, Cellular/immunology , Mesenteric Veins/immunology , Mesenteric Veins/pathology , Mesentery/blood supply
12.
Sci Rep ; 9(1): 6993, 2019 05 06.
Article in English | MEDLINE | ID: mdl-31061522

ABSTRACT

Acute-on-chronic liver disease is a clinical syndrome characterized by decompensated liver fibrosis, portal hypertension and splanchnic hyperdynamic circulation. We aimed to determine whether the alpha-1 agonist phenylephrine (Phe) facilitates endothelial nitric oxide (NO) release by mesenteric resistance arteries (MRA) in rats subjected to an experimental microsurgical obstructive liver cholestasis model (LC). Sham-operated (SO) and LC rats were maintained for eight postoperative weeks. Phe-induced vasoconstriction (in the presence/absence of the NO synthase -NOS- inhibitor L-NAME) and vasodilator response to NO donor DEA-NO were analysed. Phe-induced NO release was determined in the presence/absence of either H89 (protein kinase -PK- A inhibitor) or LY 294002 (PI3K inhibitor). PKA and PKG activities, alpha-1 adrenoceptor, endothelial NOS (eNOS), PI3K, AKT and soluble guanylate cyclase (sGC) subunit expressions, as well as eNOS and AKT phosphorylation, were determined. The results show that LC blunted Phe-induced vasoconstriction, and enhanced DEA-NO-induced vasodilation. L-NAME increased the Phe-induced contraction largely in LC animals. The Phe-induced NO release was greater in MRA from LC animals. Both H89 and LY 294002 reduced NO release in LC. Alpha-1 adrenoceptor, eNOS, PI3K and AKT expressions were unchanged, but sGC subunit expression, eNOS and AKT phosphorylation and the activities of PKA and PKG were higher in MRA from LC animals. In summary, these mechanisms may help maintaining splanchnic vasodilation and hypotension observed in decompensated LC.


Subject(s)
Acute-On-Chronic Liver Failure/drug therapy , Adrenergic alpha-1 Receptor Antagonists/pharmacology , Cyclic AMP-Dependent Protein Kinases/genetics , Liver Cirrhosis/drug therapy , Mesenteric Arteries/drug effects , Phenylephrine/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/genetics , Acute-On-Chronic Liver Failure/genetics , Acute-On-Chronic Liver Failure/metabolism , Acute-On-Chronic Liver Failure/pathology , Animals , Cholestasis/genetics , Cholestasis/metabolism , Cholestasis/pathology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinase Type I/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinase Type I/genetics , Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Hypertension, Portal/metabolism , Hypertension, Portal/pathology , Isoquinolines/pharmacology , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Male , Mesenteric Arteries/metabolism , Mesenteric Arteries/pathology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Receptors, Adrenergic, alpha-1/genetics , Receptors, Adrenergic, alpha-1/metabolism , Sulfonamides/pharmacology , Vasoconstriction/drug effects
13.
J Gastroenterol Hepatol ; 34(3): 490-494, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30345537

ABSTRACT

Hepatic encephalopathy is a severe complication of both chronic and acute liver diseases. The term hepatic encephalopathy stems from the belief that hepatic insufficiency is its fundamental etiopathogenic factor. However, most clinical cases show liver failure along with mesenteric venous portal hypertension. This portal hypertension would explain the abnormal mechanical forces suffered by the digestive tract in the early stages of the disorder. These forces could regulate some gut biochemical pathological pathways in a process known as mechanotransduction. Thus, portal hypertension would begin with the establishment of a mechanotransduced afferent or sensory inflammatory gut-brain pathway, resulting in functional and structural changes in the central nervous system. In this review, we will revisit the term "hepatic encephalopathy" in light of new results where portal hypertension occurs before liver failure and is accompanied by brain changes. Moreover, we will point out cellular links that can explain the microbiota, immune, gut, and brain axis disturbances found in this disorder.


Subject(s)
Hepatic Encephalopathy/etiology , Hypertension, Portal/complications , Liver Failure/etiology , Humans , Hypertension, Portal/physiopathology , Mechanotransduction, Cellular , Mesenteric Veins
14.
Curr Res Transl Med ; 67(2): 56-61, 2019 05.
Article in English | MEDLINE | ID: mdl-30503816

ABSTRACT

We propose that the circulatory impairments produced, in both portal hypertension and liver cirrhosis, to a certain degree resemble those characterizing prenatal life in the fetus. In fact, the left-right circulatory syndrome is common in cirrhotic patients and in the fetus. Thus, in patients with portal hypertension and chronic liver failure, the re-expression of a blood circulation comparable to fetal circulation is associated with the development of similar amniotic functions, i.e., ascites production and placenta functions, and portal vascular enteropathy. Therefore, these re-expressed embryonic functions are extra-embryonic and responsible for prenatal trophism and development.


Subject(s)
Hemodynamics/physiology , Hypertension, Portal/physiopathology , Placenta/blood supply , Placenta/physiology , Ascites/physiopathology , Female , Fetus/blood supply , Fetus/physiology , Humans , Liver Cirrhosis/physiopathology , Placental Circulation/physiology , Portal Vein/abnormalities , Portal Vein/physiopathology , Pregnancy , Splanchnic Circulation/physiology , Vascular Malformations/physiopathology
15.
Front Physiol ; 9: 1122, 2018.
Article in English | MEDLINE | ID: mdl-30174613

ABSTRACT

The aim of the present work was to study the consequences of chronic exercise training on factors involved in the regulation of mitochondrial remodeling and biogenesis, as well as the ability to produce energy and improve insulin sensitivity and glucose uptake in rat brown adipose tissue (BAT). Male Wistar rats were divided into two groups: (1) control group (C; n = 10) and (2) exercise-trained rats (ET; n = 10) for 8 weeks on a motor treadmill (five times per week for 50 min). Exercise training reduced body weight, plasma insulin, and oxidized LDL concentrations. Protein expression of ATP-independent metalloprotease (OMA1), short optic atrophy 1 (S-OPA1), and dynamin-related protein 1 (DRP1) in BAT increased in trained rats, and long optic atrophy 1 (L-OPA1) and mitofusin 1 (MFN1) expression decreased. BAT expression of nuclear respiratory factor type 1 (NRF1) and mitochondrial transcription factor A (TFAM), the main factors involved in mitochondrial biogenesis, was higher in trained rats compared to controls. Exercise training increased protein expression of sirtuin 1 (SIRT1), peroxisome proliferator-activated receptor γ coactivator 1α (PGC1α) and AMP-activated protein kinase (pAMPK/AMPK ratio) in BAT. In addition, training increased carnitine palmitoyltransferase II (CPT II), mitochondrial F1 ATP synthase α-chain, mitochondrial malate dehydrogenase 2 (mMDH) and uncoupling protein (UCP) 1,2,3 expression in BAT. Moreover, exercise increased insulin receptor (IR) ratio (IRA/IRB ratio), IRA-insulin-like growth factor 1 receptor (IGF-1R) hybrids and p42/44 activation, and decreased IGF-1R expression and IR substrate 1 (p-IRS-1) (S307) indicating higher insulin sensitivity and favoring glucose uptake in BAT in response to chronic exercise training. In summary, the present study indicates that chronic exercise is able to improve the energetic profile of BAT in terms of increased mitochondrial function and insulin sensitivity.

16.
Vascul Pharmacol ; 108: 36-45, 2018 09.
Article in English | MEDLINE | ID: mdl-29751093

ABSTRACT

We aimed to determine the influence of nitrergic innervation function on the decreased mesenteric arterial tone induced by high levels of triiodothyronine (T3), as a model of acute thyroiditis, as well as the mechanism/s implicated. We analysed in mesenteric segments from male Wistar rats the effect of 10 nmol/L T3 (2 h) on the vasomotor response to electrical field stimulation (EFS) in the presence/absence of specific neuronal NOS (nNOS) inhibitor L-NPA, or superoxide anion scavenger tempol. Nitric oxide (NO) release was measured in the presence/absence of tempol or PI3K inhibitor LY294002. Superoxide anion and peroxynitrite releases, nNOS, PI3K, AKT and superoxide dismutase (SOD) 1 and 2 expressions, nNOS and AKT phosphorylation, and SOD activity were analysed. T3 decreased EFS-induced vasoconstriction. L-NPA increased EFS-induced vasoconstriction more markedly in T3-incubated segments. T3 increased NO release. Tempol decreased EFS-induced vasoconstriction and augmented NO release only in segments without T3. LY294002 decreased NO release in T3-incubated segments. T3 diminished superoxide anion and peroxynitrite formation, enhanced SOD-2 expression, nNOS and AKT phosphorylations and SOD activity, and did not modify nNOS, PI3K, AKT and SOD-1 expressions. In conclusion, these results show a compensatory mechanism aimed at reducing the enhanced blood pressure that appears during acute thyroiditis.


Subject(s)
Mesenteric Arteries/innervation , Nitrergic Neurons/drug effects , Nitric Oxide/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Triiodothyronine/pharmacology , Vasoconstriction/drug effects , Vasodilation/drug effects , Animals , Dose-Response Relationship, Drug , Electric Stimulation , Enzyme Inhibitors/pharmacology , Free Radical Scavengers/pharmacology , In Vitro Techniques , Male , Nitrergic Neurons/enzymology , Nitric Oxide Synthase Type I/antagonists & inhibitors , Nitric Oxide Synthase Type I/metabolism , Nitrosative Stress/drug effects , Oxidative Stress/drug effects , Peroxynitrous Acid/metabolism , Phosphorylation , Rats, Wistar , Signal Transduction/drug effects , Superoxide Dismutase/metabolism , Superoxides/metabolism , Time Factors
18.
Sci Rep ; 6: 31076, 2016 08 03.
Article in English | MEDLINE | ID: mdl-27484028

ABSTRACT

We evaluated the possible alterations produced by liver cholestasis (LC), a model of decompensated liver cirrhosis in sympathetic, sensory and nitrergic nerve function in rat superior mesenteric arteries (SMA). The vasoconstrictor response to electrical field stimulation (EFS) was greater in LC animals. Alpha-adrenoceptor antagonist phentolamine and P2 purinoceptor antagonist suramin decreased this response in LC animals more than in control animals. Both non-specific nitric oxide synthase (NOS) L-NAME and calcitonin gene related peptide (CGRP) (8-37) increased the vasoconstrictor response to EFS more strongly in LC than in control segments. Vasomotor responses to noradrenaline (NA) or CGRP were greater in LC segments, while NO analogue DEA-NO induced a similar vasodilation in both experimental groups. The release of NA was not modified, while those of ATP, nitrite and CGRP were increased in segments from LC. Alpha 1 adrenoceptor, Rho kinase (ROCK) 1 and 2 and total myosin phosphatase (MYPT) expressions were not modified, while alpha 2B adrenoceptor, nNOS expression and nNOS and MYPT phosphorylation were increased by LC. Together, these alterations might counteract the increased splanchnic vasodilation observed in the last phases of decompensated liver cirrhosis.


Subject(s)
Liver Cirrhosis/physiopathology , Mesenteric Artery, Superior/physiopathology , Animals , Liver Cirrhosis/pathology , Male , Mesenteric Artery, Superior/pathology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase Type I/metabolism , Norepinephrine/pharmacology , Protein Phosphatase 1/metabolism , Rats , Rats, Wistar , Suramin/pharmacology , Vasoconstriction/drug effects , Vasodilation/drug effects
19.
PLoS One ; 11(6): e0156793, 2016.
Article in English | MEDLINE | ID: mdl-27272874

ABSTRACT

INTRODUCTION: We analysed possible time-dependent changes in nitrergic perivascular innervation function from diabetic rats and mechanisms implicated. MATERIALS AND METHODS: In endothelium-denuded mesenteric arteries from control and four- (4W) and eight-week (8W) streptozotocin-induced diabetic rats the vasoconstriction to EFS (electrical field stimulation) was analysed before and after preincubation with L-NAME. Neuronal NO release was analysed in the absence and presence of L-arginine, tetrahydrobiopterine (BH4) and L-arginine plus BH4. Superoxide anion (O2-), peroxynitrite (ONOO-) and superoxide dismutase (SOD) activity were measured. Expressions of Cu-Zn SOD, nNOS, p-nNOS Ser1417, p-nNOS Ser847, and Arginase (Arg) I and II were analysed. RESULTS: EFS response was enhanced at 4W, and to a lesser extent at 8W. L-NAME increased EFS response in control rats and at 8W, but not at 4W. NO release was decreased at 4W and restored at 8W. L-arginine or BH4 increased NO release at 4W, but not 8W. SOD activity and O2- generation were increased at both 4W and 8W. ONOO- decreased at 4W while increased at 8W. Cu-Zn SOD, nNOS and p-NOS Ser1417 expressions remained unmodified at 4W and 8W, whereas p-nNOS Ser847 was increased at 4W. ArgI was overexpressed at 4W, remaining unmodified at 8W. ArgII expression was similar in all groups. CONCLUSIONS: Our results show a time-dependent effect of diabetes on neuronal NO release. At 4W, diabetes induced increased O2- generation, nNOS uncoupling and overexpression of ArgI and p-nNOS Ser847, resulting in decreased NO release. At 8W, NO release was restored, involving normalisation of ArgI and p-nNOS Ser847 expressions.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Mesenteric Arteries/drug effects , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Vasoconstriction/drug effects , Animals , Electric Stimulation , Male , Mesenteric Arteries/metabolism , Peroxynitrous Acid/metabolism , Rats , Streptozocin , Superoxide Dismutase/metabolism , Superoxides/metabolism , Time Factors
20.
J Hypertens ; 33(9): 1819-30; discussion 1830, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26103124

ABSTRACT

INTRODUCTION: We investigated whether high-fat diet (HFD)-induced obesity was associated with modifications in mesenteric innervation function, the mechanisms involved, and the possible effects of aerobic exercise training on these changes. MATERIALS AND METHODS: Male Wistar rats were divided into three groups: rats fed a standard diet (control group); rats fed a HFD (35% fat) for 8 weeks; and HFD rats submitted to aerobic exercise training (8 weeks, 5 times per week for 50  min). Segments of isolated mesenteric arteries were exposed to electric field stimulation (EFS) with or without phentolamine, suramin, or Nω nitro-L-arginine methyl ester. Noradrenaline, ATP, and nitric oxide release, and total and phosphorylated neuronal nitric oxide synthase (nNOS, P-nNOS) expression were also measured. RESULTS: EFS contraction was greater in sedentary HFD than in control rats. Phentolamine reduced EFS contractions more markedly in HFD rats. Suramin decreased EFS contractions only in control rats. Phentolamine + suramin practically abolished EFS-induced contraction in control rats, whereas it did not modify it in the HFD rats. Noradrenaline release was greater and ATP was lower in HFD rats. Nω nitro-L-arginine methyl ester increased contractions to EFS only in segments from control rats. Nitric oxide release and nNOS and P-nNOS expressions were lower in arterial segments from HFD rats than from control rats. None of these changes in sedentary HFD rats was present in the trained HFD rats. CONCLUSIONS: Enhanced sympathetic and diminished nitrergic components contributed to increased vasoconstrictor responses to EFS in sedentary HFD rats. All these changes were avoided by aerobic exercise training, suggesting that aerobic exercise could reduce peripheral vascular resistance in obesity.


Subject(s)
Mesenteric Arteries/physiopathology , Nitrergic Neurons/physiology , Obesity/physiopathology , Physical Conditioning, Animal/physiology , Sympathetic Nervous System/physiopathology , Adenosine Triphosphate/metabolism , Animals , Antihypertensive Agents/pharmacology , Diet, High-Fat , Electric Stimulation , Enzyme Inhibitors/pharmacology , Male , Mesenteric Arteries/drug effects , Mesenteric Arteries/innervation , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type I/metabolism , Norepinephrine/metabolism , Phentolamine/pharmacology , Rats , Rats, Wistar , Suramin/pharmacology , Vasoconstriction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...