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










Database
Publication year range
1.
Biomed Pharmacother ; 174: 116505, 2024 May.
Article in English | MEDLINE | ID: mdl-38574614

ABSTRACT

Pulmonary arterial hypertension (PAH) was a devastating disease characterized by artery remodeling, ultimately resulting in right heart failure. The aim of this study was to investigate the effects of canagliflozin (CANA), a sodium-glucose cotransporter 2 inhibitor (SGLT2i) with mild SGLT1 inhibitory effects, on rats with PAH, as well as its direct impact on pulmonary arterial smooth muscle cells (PASMCs). PAH rats were induced by injection of monocrotaline (MCT) (40 mg/kg), followed by four weeks of treatment with CANA (30 mg/kg/day) or saline alone. Pulmonary artery and right ventricular (RV) remodeling and dysfunction in PAH were alleviated with CANA, as assessed by echocardiography. Hemodynamic parameters and structural of pulmonary arteriole, including vascular wall thickness and wall area, were reduced by CANA. RV hypertrophy index, cardiomyocyte hypertrophy, and fibrosis were decreased with CANA treatment. PASMCs proliferation was inhibited by CANA under stimulation by platelet-derived growth factor (PDGF)-BB or hypoxia. Activation of AMP kinase (AMPK) was induced by CANA treatment in cultured PASMCs in a time- and concentration-dependent manner. These effects of CANA were attenuated when treatment with compound C, an AMPK inhibitor. Abundant expression of SGLT1 was observed in PASMCs and pulmonary arteries, while SGLT2 expression was undetectable. SGLT1 increased in response to PDGF-BB or hypoxia stimulation, while PASMCs proliferation was inhibited and beneficial effects of CANA were counteracted by knockdown of SGLT1. Our research demonstrated for the first time that CANA inhibited the proliferation of PASMCs by regulating SGLT1/AMPK signaling and thus exerted an anti-proliferative effect on MCT-induced PAH.


Subject(s)
Canagliflozin , Cell Proliferation , Myocytes, Smooth Muscle , Pulmonary Arterial Hypertension , Vascular Remodeling , Animals , Rats , AMP-Activated Protein Kinases/drug effects , AMP-Activated Protein Kinases/metabolism , Canagliflozin/pharmacology , Cell Proliferation/drug effects , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/metabolism , Monocrotaline/adverse effects , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Myocytes, Smooth Muscle/metabolism , Pulmonary Arterial Hypertension/drug therapy , Pulmonary Arterial Hypertension/pathology , Pulmonary Arterial Hypertension/metabolism , Pulmonary Arterial Hypertension/chemically induced , Pulmonary Artery/drug effects , Pulmonary Artery/pathology , Pulmonary Artery/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , Sodium-Glucose Transporter 1/drug effects , Sodium-Glucose Transporter 1/metabolism , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Vascular Remodeling/drug effects
2.
Vet Med Sci ; 5(3): 451-461, 2019 08.
Article in English | MEDLINE | ID: mdl-30973212

ABSTRACT

The ban on the use of antibiotic in feed encouraged nutritionists to using alternatives to maintain growth performance and intestinal function of broilers. This study was conducted to evaluate the effects of Yupingfeng polysaccharides (YP) supplementation on growth performance and expression of SGLT1, GLUT2 and GLUT5 in Qingyuan partridge chicken. Experiment 1: a total of 540 chickens were randomly allocated to five groups with six replication. Dietary treatments were: (1) CON (control group), basal diet; (2) T1, CON + 0.5 g kg-1 YP; (3) T2, CON + 1 g kg-1 YP; (4) T3, CON + 2 g kg-1 YP; (5) T4, CON + 4 g kg-1 YP. Experiment 2, a total of 162 were randomly allocated to three groups with three replication. Dietary treatments were: (1) CON, basal diet; (2) T1, CON + 0.5 g kg-1 YP; (3) T2, CON + 1 g kg-1 YP. From days 1 to 14 and overall, chicken fed T1 diet had higher ADG. On day 42, there was increased villus height of jejunum in T1 group. On days 14 and 28, there was decreased villus height of duodenum and jejunum in T2 group. In duodenum, the expression of SGLT1 (days 21, 35 and 42), GLUT2 (days 7, 14, 21, 28, 35 and 42) and GLUT5 (days 7, 14, 21 and 28) was increased with YP supplementation. In jejunum, the expression of SGLT1 (days 7, 14, 21, 28 and 35), GLUT2 (days 14, 21, 28, 35 and 42) and GLUT5 (days 7, 14, 21, 28, 35 and 42) was increased with YP supplementation. In ileum, the expression of SGLT1 (days 7, 21, 35 and 42), GLUT2 (days 7, 14, 21 and 42) and GLUT5 (days 7, 14, 21, 28, 35 and 42) was increased with YP supplementation. Dietary YP supplementation improves growth performance and expression of SGLT1, GLUT2 and GLUT5 in intestine.


Subject(s)
Chickens/growth & development , Dietary Supplements/analysis , Drugs, Chinese Herbal/pharmacology , Gene Expression Regulation, Developmental/drug effects , Polysaccharides/pharmacology , Animal Feed/analysis , Animals , Chickens/anatomy & histology , Chickens/genetics , Diet/veterinary , Gene Expression Regulation, Developmental/genetics , Glucose Transporter Type 2/drug effects , Glucose Transporter Type 2/genetics , Glucose Transporter Type 5/drug effects , Glucose Transporter Type 5/genetics , Intestinal Mucosa/anatomy & histology , Intestinal Mucosa/drug effects , Intestine, Small/anatomy & histology , Intestine, Small/drug effects , RNA, Messenger/drug effects , RNA, Messenger/genetics , Random Allocation , Sodium-Glucose Transporter 1/drug effects , Sodium-Glucose Transporter 1/genetics , Up-Regulation
3.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 45(6): 598-606, 2016 05 25.
Article in Chinese | MEDLINE | ID: mdl-28247603

ABSTRACT

Objective: To investigate the role of glucose transporter 1 (GLUT1) and sodium-glucose cotransporter 1 (SGLT1) in high glucose dialysate-induced peritoneal fibrosis. Methods: Thirty six male SD rats were randomly divided into 6 groups (6 in each):normal control group, sham operation group, peritoneal dialysis group (PD group), PD+phloretin group (PD+T group), PD+phlorizin group (PD+Z group), PD+phloretin+phlorizin group (PD+T+Z group). Rat model of uraemia was established using 5/6 nephrotomy, and 2.5% dextrose peritoneal dialysis solution was used in peritoneal dialysis. Peritoneal equilibration test was performed 24 h after dialysis to evaluate transport function of peritoneum in rats; HE staining was used to observe the morphology of peritoneal tissue; and immunohistochemistry was used to detect the expression of GLUT1, SGLT1, TGF-ß1 and connective tissue growth factor (CTGF) in peritoneum. Human peritoneal microvascular endothelial cells (HPECs) were divided into 5 groups:normal control group, peritoneal dialysis group (PD group), PD+phloretin group (PD+T group), PD+phlorezin group (PD+Z group), and PD+phloretin+phlorezin group (PD+T+Z group). Real time PCR and Western blotting were used to detect mRNA and protein expressions of GLUT1, SGLT1, TGF-ß1, CTGF in peritoneal membrane and HPECs. Results:In vivo, compared with sham operation group, rats in PD group had thickened peritoneum, higher ultrafiltration volume, and the mRNA and protein expressions of GLUT1, SGLT1, CTGF, TGF-ß1 were significantly increased (all P<0.05); compared with PD group, thickened peritoneum was attenuated, and the mRNA and protein expressions of GLUT1, SGLT1, CTGF, TGF-ß1 were significantly decreased in PD+T, PD+Z and PD+T+Z groups (all P<0.05). Pearson's correlation analysis showed that the expressions of GLUT1, SGLT1 in peritoneum were positively correlated with the expressions of TGF-ß1 and CTGF (all P<0.05). In vitro, the mRNA and protein expressions of GLUT1, SGLT1, TGF-ß1, CTGF were significantly increased in HPECs of peritoneal dialysis group (all P<0.05), and those in PD+T, PD+Z, and PD+T+Z groups were decreased (all P<0.05). Pearson's correlation analysis showed that the expressions of GLUT1, SGLT1 in HPECs were positively correlated with the expressions of TGF-ß1 and CTGF (all P<0.05). Conclusion: High glucose peritoneal dialysis fluid may promote peritoneal fibrosis by upregulating the expressions of GLUT1 and SGLT1.


Subject(s)
Dialysis Solutions/adverse effects , Dialysis Solutions/pharmacology , Glucose Transporter Type 1/drug effects , Glucose Transporter Type 1/physiology , Glucose/adverse effects , Glucose/pharmacology , Hemodiafiltration/adverse effects , Hemodiafiltration/methods , Peritoneal Dialysis/adverse effects , Peritoneal Dialysis/methods , Peritoneal Fibrosis/chemically induced , Peritoneal Fibrosis/genetics , Peritoneum/chemistry , Peritoneum/drug effects , Peritoneum/pathology , Sodium-Glucose Transporter 1/drug effects , Sodium-Glucose Transporter 1/physiology , Uremia/chemically induced , Animals , Cells, Cultured , Connective Tissue Growth Factor/analysis , Connective Tissue Growth Factor/drug effects , Dialysis Solutions/chemistry , Gene Expression Regulation/drug effects , Glucose Transporter Type 1/analysis , Humans , Male , Peritoneal Fibrosis/physiopathology , Phloretin , Phlorhizin , RNA, Messenger , Rats , Rats, Sprague-Dawley , Sodium-Glucose Transporter 1/analysis , Transforming Growth Factor beta1/analysis , Transforming Growth Factor beta1/drug effects
4.
Am J Physiol Renal Physiol ; 306(12): F1520-33, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24761001

ABSTRACT

Sodium-glucose cotransporter 2 (SGLT2) inhibitors showed a glucose lowering effect in type 2 diabetes patients through inducing renal glucose excretion. Detailed analysis of the mechanism of the glucosuric effect of SGLT2 inhibition, however, has been hampered by limitations of clinical study. Here, we investigated the mechanism of urinary glucose excretion using nonhuman primates with SGLT inhibitors tofogliflozin and phlorizin, both in vitro and in vivo. In cells overexpressing cynomolgus monkey SGLT2 (cSGLT2), both tofogliflozin and phlorizin competitively inhibited uptake of the substrate (α-methyl-d-glucopyranoside; AMG). Tofogliflozin was found to be a selective cSGLT2 inhibitor, inhibiting cSGLT2 more strongly than did phlorizin, with selectivity toward cSGLT2 1,000 times that toward cSGLT1; phlorizin was found to be a nonselective cSGLT1/2 inhibitor. In a glucose titration study in cynomolgus monkeys under conditions of controlled plasma drug concentration, both tofogliflozin and phlorizin increased fractional excretion of glucose (FEG) by up to 50% under hyperglycemic conditions. By fitting the titration curve using a newly introduced method that avoids variability in estimating the threshold of renal glucose excretion, we found that tofogliflozin and phlorizin lowered the threshold and extended the splay in a dose-dependent manner without significantly affecting the tubular transport maximum for glucose (TmG). Our results demonstrate the contribution of SGLT2 to renal glucose reabsorption (RGR) in cynomolgus monkeys and demonstrate that competitive inhibition of cSGLT2 exerts a glucosuric effect by mainly extending splay and lowering threshold without affecting TmG.


Subject(s)
Benzhydryl Compounds/pharmacology , Glucose/metabolism , Glucosides/pharmacology , Macaca fascicularis/urine , Phlorhizin/pharmacology , Sodium-Glucose Transporter 2 Inhibitors , Sodium-Glucose Transporter 2/drug effects , Animals , COS Cells/metabolism , COS Cells/pathology , Chlorocebus aethiops , DNA, Complementary/genetics , Dose-Response Relationship, Drug , In Vitro Techniques , Kidney/metabolism , Kidney/pathology , Male , Methylglucosides/metabolism , Models, Animal , Sodium-Glucose Transporter 1/antagonists & inhibitors , Sodium-Glucose Transporter 1/drug effects , Sodium-Glucose Transporter 1/genetics , Sodium-Glucose Transporter 2/genetics
5.
Am J Physiol Regul Integr Comp Physiol ; 305(7): R840-53, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23926132

ABSTRACT

Post-oral sugar actions enhance the intake of and preference for sugar-rich foods, a process referred to as appetition. Here, we investigated the role of intestinal sodium glucose cotransporters (SGLTs) in sugar appetition in C57BL/6J mice using sugars and nonmetabolizable sugar analogs that differ in their affinity for SGLT1 and SGLT3. In experiments 1 and 2, food-restricted mice were trained (1 h/day) to consume a flavored saccharin solution [conditioned stimulus (CS-)] paired with intragastric (IG) self-infusions of water and a different flavored solution (CS+) paired with infusions of 8 or 12% sugars (glucose, fructose, and galactose) or sugar analogs (α-methyl-D-glucopyranoside, MDG; 3-O-methyl-D-glucopyranoside, OMG). Subsequent two-bottle CS+ vs. CS- choice tests were conducted without coinfusions. Infusions of the SGLT1 ligands glucose, galactose, MDG, and OMG stimulated CS+ licking above CS- levels. However, only glucose, MDG, and galactose conditioned significant CS+ preferences, with the SGLT3 ligands (glucose, MDG) producing the strongest preferences. Fructose, which is not a ligand for SGLTs, failed to stimulate CS+ intake or preference. Experiment 3 revealed that IG infusion of MDG+phloridzin (an SGLT1/3 antagonist) blocked MDG appetition, whereas phloridzin had minimal effects on glucose-induced appetition. However, adding phloretin (a GLUT2 antagonist) to the glucose+phloridzin infusion blocked glucose appetition. Taken together, these findings suggest that humoral signals generated by intestinal SGLT1 and SGLT3, and to a lesser degree, GLUT2, mediate post-oral sugar appetition in mice. The MDG results indicate that sugar metabolism is not essential for the post-oral intake-stimulating and preference-conditioning actions of sugars in mice.


Subject(s)
Appetite Regulation/drug effects , Carbohydrates/administration & dosage , Food Preferences/drug effects , Administration, Oral , Animals , Conditioning, Psychological/drug effects , Drinking/drug effects , Fructose/administration & dosage , Fructose/metabolism , Galactose/administration & dosage , Galactose/metabolism , Glucose/administration & dosage , Glucose/metabolism , Glucose Transporter Type 2/drug effects , Glucose Transporter Type 2/metabolism , Intestinal Absorption , Intestinal Mucosa/metabolism , Intestines/drug effects , Ligands , Male , Methylglucosides/administration & dosage , Methylglucosides/metabolism , Mice , Mice, Inbred C57BL , Saccharin/administration & dosage , Self Administration , Sodium-Glucose Transport Proteins/drug effects , Sodium-Glucose Transport Proteins/metabolism , Sodium-Glucose Transporter 1/drug effects , Sodium-Glucose Transporter 1/metabolism , Sweetening Agents/administration & dosage , Time Factors
6.
J Chem Inf Model ; 53(3): 661-73, 2013 Mar 25.
Article in English | MEDLINE | ID: mdl-23351136

ABSTRACT

Traditional Chinese medicine (TCM) and Ayurveda have been used in humans for thousands of years. While the link to a particular indication has been established in man, the mode-of-action (MOA) of the formulations often remains unknown. In this study, we aim to understand the MOA of formulations used in traditional medicine using an in silico target prediction algorithm, which aims to predict protein targets (and hence MOAs), given the chemical structure of a compound. Following this approach we were able to establish several links between suggested MOAs and experimental evidence. In particular, compounds from the 'tonifying and replenishing medicinal' class from TCM exhibit a hypoglycemic effect which can be related to activity of the ingredients against the Sodium-Glucose Transporters (SGLT) 1 and 2 as well as Protein Tyrosine Phosphatase (PTP). Similar results were obtained for Ayurvedic anticancer drugs. Here, both primary anticancer targets (those directly involved in cancer pathogenesis) such as steroid-5-alpha-reductase 1 and 2 were predicted as well as targets which act synergistically with the primary target, such as the efflux pump P-glycoprotein (P-gp). In addition, we were able to elucidate some targets which may point us to novel MOAs as well as explain side effects. Most notably, GPBAR1, which was predicted as a target for both 'tonifying and replenishing medicinal' and anticancer classes, suggests an influence of the compounds on metabolism. Understanding the MOA of these compounds is beneficial as it provides a resource for NMEs with possibly higher efficacy in the clinic than those identified by single-target biochemical assays.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Medicine, Ayurvedic , Medicine, Chinese Traditional , ATP Binding Cassette Transporter, Subfamily B, Member 1/drug effects , Algorithms , Antineoplastic Agents/pharmacology , Artificial Intelligence , Computer Simulation , Databases, Genetic , Humans , Hypoglycemic Agents/pharmacology , Plants, Medicinal/chemistry , Plants, Medicinal/genetics , Protein Tyrosine Phosphatases/drug effects , Receptors, G-Protein-Coupled/drug effects , Sodium-Glucose Transporter 1/drug effects , Sodium-Glucose Transporter 2/drug effects
7.
Chem Res Toxicol ; 23(2): 405-12, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20073493

ABSTRACT

Cadmium is a major environmental pollutant that causes kidney failure including the inability to resorb nutrients such as glucose. In a mouse kidney cell culture model, Cd(2+) inhibits Na(+)-dependent glucose uptake mediated by SGLT transporters. This defect has been traced to the down-regulation of SGLT mRNA synthesis mediated by the zinc-finger transcription factor, Zn(3)-Sp1. Incubation of Cd(2+) with Zn(2+)-Sp1 inhibited its capacity to bind to GC1, its binding site in the SGLT1 promoter. The extent of reaction was reduced as increasing concentrations of Zn(2+) are simultaneously present in the reaction mixture. The results are consistent with a Cd(2+)-Zn(2+) exchange reaction that inactivates the DNA binding function of the protein. The equilibrium constant for this reaction was calculated as 14 +/- 3 and 7 +/- 4 for the reactions measured by the binding to GC1 and an analogous SGLT2 promoter site. Sequential addition of Cd(2+) and Zn(2+) to Zn(3)-Sp1 failed to inhibit the reduction in DNA binding seen with Cd(2+) alone, indicating that substitution of Zn(2+) by Cd(2+) was followed by a second reaction that failed to respond to Zn(2+). Buffers for the DNA binding reaction (electrophoretic mobility shift assay) contain EDTA and Cd-EDTA is active in the same concentration range as Cd(2+). During the standard 15 min incubation, Cd(2+) down-regulates Zn(3)-Sp1 but is inactive against the adduct, Zn(3)-Sp1.GC1. Kinetic studies demonstrated that with 5 muM Cd(2+), Zn(3)-Sp1 was about 75% inactivated in 15 min, whereas, Zn(3)-Sp1.GC1 was slowly dissociated with 50% still remaining after 60 min. In contrast, Zn(3)-Sp1 bound to a cognate consensus site resisted any reaction over 60 min. An adduct of Zn(3)-Sp1.(polydI-dC) was just as reactive with Cd(2+) as Zn(3)-Sp1. Reexamination of the NMR structure of Zn- and Cd-finger peptides related to Sp1 fingers has revealed subtle changes in conformation of the metalbinding site and DNA-binding helix that occur when Cd(2+) is substituted by Zn(2+).


Subject(s)
Cadmium/toxicity , Transcription Factors/chemistry , Zinc Fingers , Animals , Base Sequence , Binding, Competitive , Cadmium/chemistry , Cell Line , Consensus Sequence , Down-Regulation/drug effects , Kidney/cytology , Mice , Molecular Sequence Data , Sodium-Glucose Transporter 1/drug effects , Sodium-Glucose Transporter 1/metabolism , Transcription Factors/drug effects , Zinc Fingers/drug effects
8.
Biophys Chem ; 127(1-2): 69-77, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17222499

ABSTRACT

Recombinant purified human sodium/D-glucose cotransporter1 (hSGLT1) was reconstituted in a functional form into phospholipid vesicles and its conformational states in the absence and presence of ligands and inhibitors were probed by intrinsic tryptophan fluorescence. In the presence of sodium, sugars increase intrinsic fluorescence (maximum 17%) in a saturable manner in the following order alpha-MDG >D-Glu approximately D-Gal >> D-Man >D-All, with no effect of L-Glu. Apparent affinities ranging from 0.65 to 10.4 mM were observed. In addition, D-Glu increased the accessibility of the Trps to hydrophilic collisional quenchers. On the contrary, the transport inhibitor phlorizin decreased Trps fluorescence in a sodium-dependent manner by 50% with a red shift of 4-6 nm and decreased quencher accessibility, these effects were saturable with a high affinity of 5 microM. Furthermore, the positioning of the tryptophans in the reconstituted transporter was investigated. hSGLT1 Trps fluorescence was reduced by N-bromosuccinimide treatment maximally 25% in membranes and 65% in solution. The fluorescence was also significantly but differently quenched by the lipid-soluble spin labeled probes 5-Doxyl-phosphatidylcholine (40%) and 12-Doxyl-phosphatidylcholine (26%). Depth-calculation using the parallax method suggested a location of Trps at an average depth of 10 angstrom from the center of the bilayer. These studies demonstrate the existence of different conformational states of the membrane-embedded transporter in its glucose-free form, as sodium-glucose-carrier complex and as sodium-phlorizin-carrier complex. They further indicate that most of the Trp residues in hSGLT1 are located in hydrophobic regions of the protein or in contact with the lipid bilayer of the membrane. There, they are located close to the membrane-water interface contributing to the vectorial nature of the transporter.


Subject(s)
Sodium-Glucose Transporter 1/chemistry , Tryptophan/analysis , Cyclic N-Oxides/chemistry , Fluorescence , Humans , Ligands , Liposomes/chemistry , Membranes/chemistry , Phlorhizin/pharmacology , Phospholipids/chemistry , Protein Conformation , Sodium-Glucose Transporter 1/drug effects , Sodium-Glucose Transporter 1/metabolism , Spin Labels
SELECTION OF CITATIONS
SEARCH DETAIL
...