Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Cell Biol Int ; 2024 May 13.
Article in English | MEDLINE | ID: mdl-38741282

ABSTRACT

Polycystic ovary syndrome (PCOS) is the primary cause of female infertility with a lack of universal therapeutic regimen. Although osthole exhibits numerous pharmacological activities in treating various diseases, its therapeutic effect on PCOS is undiscovered. The present study found that application of osthole improved the symptoms of PCOS mice through preventing ovarian granulosa cells (GCs) production of more estrogen and alleviating the liberation of pro-inflammatory cytokine interleukin (IL)-1ß, IL-6, and tumor necrosis factor alpha. Meanwhile, osthole enhanced ovarian antioxidant capacity and alleviated intracellular reactive oxygen species (ROS) accumulation with a concurrent attenuation for oxidative stress, while intervention of antioxidant enzymic activity and glutathione (GSH) synthesis neutralized the salvation of osthole on GCs secretory disorder and chronic inflammation. Further analysis revealed that osthole restored the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and forkhead box O 1 (Foxo1) whose repression antagonized the amelioration of osthole on the insufficiency of antioxidant capacity and accumulation of ROS. Moreover, Nrf2 served as an intermedium to mediate the regulation of osthole on Foxo1. Additionally, osthole restricted the phosphorylation of IκBα and nuclear factor kappa B (NF-κB) subunit p65 by DHEA and weakened the transcriptional activity of NF-κB, but this effectiveness was abrogated by the obstruction of Nrf2 and Foxo1, whereas adjunction of GSH renewed the redemptive effect of osthole on NF-κB whose activation caused an invalidation of osthole in rescuing the aberration of GCs secretory function and inflammation response. Collectively, osthole might relieve the symptoms of PCOS mice via Nrf2-Foxo1-GSH-NF-κB pathway.

2.
J Agric Food Chem ; 72(6): 2963-2976, 2024 Feb 14.
Article in English | MEDLINE | ID: mdl-38305024

ABSTRACT

Polycystic ovarian syndrome (PCOS) is the major cause of infertility in reproductive women, but no universal drug is feasible. Although puerarin clinically treats cerebrovascular and cardiovascular diseases, its curative effect on PCOS remains elusive. The present study discovered that administration of puerarin restored estrous cycle of PCOS mice and diminished the number of cystic follicles with the concomitant recovery for circulating testosterone, LH and FSH levels, and LH/FSH ratio, indicating the therapeutic role of puerarin in PCOS. KEGG analysis of differential genes between PCOS and control revealed the enrichment in MAPK and calcium signaling pathway. Application of puerarin restricted the phosphorylation of ERK1/2 and JNK, whose activation neutralized the improvement of puerarin on the secretory function and apoptosis of ovarian granulosa cells (GCs). Meanwhile, puerarin alleviated the accumulation of cytosolic Ca2+ through restricting the opening of Ryr and Itpr channels, but this effectiveness was counteracted by the activatory ERK1/2 and JNK. Attenuation of cytosolic Ca2+ counteracted the antagonistic effects of ERK1/2 and JNK activation on puerarin's role in rescuing the calcineurin and Nfatc. Further analysis manifested that Mcu had been authenticated as a direct downstream target of Nfatc to mediate the amelioration of puerarin on mitochondrial Ca2+ uptake. Moreover, puerarin prevented the disorder of ATP content, mitochondrial membrane potential, and mitochondrial permeability transition pore opening through maintaining mitochondrial Ca2+ homeostasis. Collectively, puerarin might ameliorate the symptoms of PCOS mice through preventing mitochondrial dysfunction that is dependent on the maintenance of intracellular Ca2+ homeostasis after inactivation of ERK1/2 and JNK.


Subject(s)
Isoflavones , Mitochondrial Diseases , Polycystic Ovary Syndrome , Female , Humans , Mice , Animals , Polycystic Ovary Syndrome/drug therapy , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/metabolism , Calcium/metabolism , Granulosa Cells , Follicle Stimulating Hormone/metabolism , Follicle Stimulating Hormone/pharmacology , Follicle Stimulating Hormone/therapeutic use , Mitochondrial Diseases/metabolism
3.
Proc Natl Acad Sci U S A ; 120(33): e2305717120, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37549287

ABSTRACT

Great progress has been made in identifying positive regulators that activate adipocyte thermogenesis, but negative regulatory signaling of thermogenesis remains poorly understood. Here, we found that cardiotrophin-like cytokine factor 1 (CLCF1) signaling led to loss of brown fat identity, which impaired thermogenic capacity. CLCF1 levels decreased during thermogenic stimulation but were considerably increased in obesity. Adipocyte-specific CLCF1 transgenic (CLCF1-ATG) mice showed impaired energy expenditure and severe cold intolerance. Elevated CLCF1 triggered whitening of brown adipose tissue by suppressing mitochondrial biogenesis. Mechanistically, CLCF1 bound and activated ciliary neurotrophic factor receptor (CNTFR) and augmented signal transducer and activator of transcription 3 (STAT3) signaling. STAT3 transcriptionally inhibited both peroxisome proliferator-activated receptor-γ coactivator (PGC) 1α and 1ß, which thereafter restrained mitochondrial biogenesis in adipocytes. Inhibition of CNTFR or STAT3 could diminish the inhibitory effects of CLCF1 on mitochondrial biogenesis and thermogenesis. As a result, CLCF1-TG mice were predisposed to develop metabolic dysfunction even without external metabolic stress. Our findings revealed a brake signal on nonshivering thermogenesis and suggested that targeting this pathway could be used to restore brown fat activity and systemic metabolic homeostasis in obesity.


Subject(s)
Adipocytes, Brown , Organelle Biogenesis , Animals , Mice , Adipocytes, Brown/metabolism , Adipose Tissue, Brown/metabolism , Homeostasis , Obesity/genetics , Obesity/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Thermogenesis/physiology
4.
Cell Rep ; 42(7): 112731, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37393616

ABSTRACT

Energy-dissipating adipocytes have the potential to improve metabolic health. Here, we identify hypoxia-induced gene domain protein-1a (HIGD1A), a mitochondrial inner membrane protein, as a positive regulator of adipose browning. HIGD1A is induced in thermogenic fats by cold exposure. Peroxisome proliferator-activated receptor gamma (PPARγ) transactivates HIGD1A expression synergistically with peroxisome proliferators-activated receptor γ coactivator α (PGC1α). HIGD1A knockdown inhibits adipocyte browning, whereas HIGD1A upregulation promotes the browning process. Mechanistically, HIGD1A deficiency impairs mitochondrial respiration to increase reactive oxygen species (ROS) level. This increases NAD+ consumption for DNA damage repair and curtails the NAD+/NADH ratio, which inhibits sirtuin1 (SIRT1) activity, thereby compromising adipocyte browning. Conversely, overexpression of HIGD1A blunts the above process to promote adaptive thermogenesis. Furthermore, mice with HIGD1A knockdown in inguinal and brown fat have impaired thermogenesis and are prone to diet-induced obesity (DIO). Overexpression of HIGD1A favors adipose tissue browning, ultimately preventing DIO and metabolic disorders. Thus, the mitochondrial protein HIGD1A links SIRT1 activity to adipocyte browning by inhibiting ROS levels.


Subject(s)
NAD , Sirtuin 1 , Animals , Mice , Adipocytes, Brown/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , DNA Damage , Mice, Inbred C57BL , NAD/metabolism , Obesity/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Reactive Oxygen Species/metabolism , Sirtuin 1/genetics , Sirtuin 1/metabolism , Thermogenesis/genetics
5.
Nat Metab ; 4(10): 1352-1368, 2022 10.
Article in English | MEDLINE | ID: mdl-36253617

ABSTRACT

Cysteine dioxygenase 1 (Cdo1) is a key enzyme in taurine synthesis. Here we show that Cdo1 promotes lipolysis in adipose tissue. Adipose-specific knockout of Cdo1 in mice impairs energy expenditure, cold tolerance and lipolysis, exacerbates diet-induced obesity (DIO) and decreases adipose expression of the key lipolytic genes encoding ATGL and HSL, with little effect on adipose taurine levels. White-adipose-specific overexpression of ATGL and HSL blunts the role of adipose Cdo1 deficiency in promoting DIO. Mechanistically, Cdo1 interacts with PPARγ and facilitates the recruitment of Med24, the core subunit of mediator complex, to ATGL and HSL gene promoters, thereby transactivating their expression. Further, mice with transgenic overexpression of Cdo1 show better cold tolerance, ameliorated DIO and higher lipolysis capacity. Thus, we uncover an unexpected and important role of Cdo1 in regulating adipose lipolysis.


Subject(s)
Lipolysis , PPAR gamma , Male , Mice , Animals , Lipolysis/physiology , PPAR gamma/genetics , PPAR gamma/metabolism , Cysteine Dioxygenase/metabolism , Sterol Esterase/metabolism , Lipase/metabolism , Adipose Tissue/metabolism , Obesity/genetics , Obesity/metabolism , Mediator Complex/metabolism , Taurine/metabolism
6.
Mol Metab ; 61: 101500, 2022 07.
Article in English | MEDLINE | ID: mdl-35436587

ABSTRACT

OBJECTIVE: Due to the increasing prevalence of obesity and insulin resistance, there is an urgent need for better treatment of obesity and its related metabolic disorders. This study aimed to elucidate the role of SERPINA3C, an adipocyte secreted protein, in obesity and related metabolic disorders. METHODS: Male wild type (WT) and knockout (KO) mice were fed with high-fat diet (HFD) for 16 weeks, adiposity, insulin resistance, and inflammation were assessed. AAV-mediated overexpression of SERPINA3C was injected locally in inguinal white adipose tissue (iWAT) to examine the effect of SERPINA3C. In vitro analyses were conducted in 3T3-L1 adipocytes to explore the molecular pathways underlying the function of SERPINA3C. RESULTS: Functional exploration of the SERPINA3C knockout mice revealed that SERPINA3C deficiency led to an impaired metabolic phenotype (more severe obesity, lower metabolic rates, worse glucose intolerance and insulin insensitivity), which was associated with anabatic inflammation and apoptosis of white adipose tissues. Consistent with these results, overexpression of SERPINA3C in inguinal adipose tissue protected mice against diet-induced obesity and metabolic disorders with less inflammation and apoptosis in adipose tissue. Mechanistically, SERPINA3C inhibited Cathepsin G activity, acting as a serine protease inhibitor, which blocked Cathepsin G-mediated turnover of α5/ß1 Integrin protein. Then, the preserved integrity (increase) of α5/ß1 Integrin signaling activated AKT to decrease JNK phosphorylation, thereby inhibiting inflammation and promoting insulin sensitivity in adipocytes. CONCLUSIONS/INTERPRETATION: These findings demonstrate a previously unknown SERPINA3C/Cathepsin G/Integrin/AKT pathway in regulating adipose tissue inflammation, and suggest the therapeutic potential of targeting SERPINA3C/Cathepsin G axis in adipose tissue for the treatment of obesity and metabolic diseases.


Subject(s)
Adipose Tissue , Cathepsin G , Insulin Resistance , Integrin alpha5beta1 , Obesity , Serpins , Adipocytes/metabolism , Adipose Tissue/metabolism , Animals , Cathepsin G/metabolism , Cathepsin G/pharmacology , Diet, High-Fat/adverse effects , Inflammation/drug therapy , Inflammation/metabolism , Insulin Resistance/physiology , Integrin alpha5beta1/metabolism , Integrin beta1/metabolism , Integrins/metabolism , Male , Mice , Mice, Knockout , Obesity/etiology , Obesity/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Serpins/deficiency , Serpins/metabolism
7.
Diabetes ; 70(7): 1458-1472, 2021 07.
Article in English | MEDLINE | ID: mdl-33863801

ABSTRACT

l-Theanine is a nonprotein amino acid with much beneficial efficacy. We found that intraperitoneal treatment of the mice with l-theanine (100 mg/kg/day) enhanced adaptive thermogenesis and induced the browning of inguinal white adipose tissue (iWAT) with elevated expression of Prdm16, Ucp1, and other thermogenic genes. Meanwhile, administration of the mice with l-theanine increased energy expenditure. In vitro studies indicated that l-theanine induced the development of brown-like features in adipocytes. The shRNA-mediated depletion of Prdm16 blunted the role of l-theanine in promoting the brown-like phenotypes in adipocytes and in the iWAT of mice. l-theanine treatment enhanced AMPKα phosphorylation both in adipocytes and iWAT. Knockdown of AMPKα abolished l-theanine-induced upregulation of Prdm16 and adipocyte browning. l-Theanine increased the α-ketoglutarate (α-KG) level in adipocytes, which may increase the transcription of Prdm16 by inducing active DNA demethylation on its promoter. AMPK activation was required for l-theanine-induced increase of α-KG and DNA demethylation on the Prdm16 promoter. Moreover, intraperitoneal administration with l-theanine ameliorated obesity, improved glucose tolerance and insulin sensitivity, and reduced plasma triglyceride, total cholesterol, and free fatty acids in the high-fat diet-fed mice. Our results suggest a potential role of l-theanine in combating diet-induced obesity in mice, which may involve l-theanine-induced browning of WAT.


Subject(s)
AMP-Activated Protein Kinases/physiology , Adipose Tissue, White/drug effects , DNA-Binding Proteins/physiology , Glutamates/pharmacology , Ketoglutaric Acids/metabolism , Maillard Reaction/drug effects , Obesity/prevention & control , Transcription Factors/physiology , Adipose Tissue, White/metabolism , Animals , DNA Methylation , DNA-Binding Proteins/genetics , Diet, High-Fat , Male , Mice, Inbred C57BL , Transcription Factors/genetics
8.
J Biol Chem ; 294(41): 15014-15024, 2019 10 11.
Article in English | MEDLINE | ID: mdl-31427436

ABSTRACT

Taurine, a nonprotein amino acid, is widely distributed in almost all animal tissues. Ingestion of taurine helps to improve obesity and its related metabolic disorders. However, the molecular mechanism underlying the protective role of taurine against obesity is not completely understood. In this study, it was found that intraperitoneal treatment of mice with taurine alleviated high-fat diet (HFD)-induced obesity, improved insulin sensitivity, and increased energy expenditure and adaptive thermogenesis of the mice. Meanwhile, administration of the mice with taurine markedly induced the browning of inguinal white adipose tissue (iWAT) with significantly elevated expression of PGC1α, UCP1, and other thermogenic genes in iWAT. In vitro studies indicated that taurine also induced the development of brown-like adipocytes in C3H10T1/2 white adipocytes. Knockdown of PGC1α blunted the role of taurine in promoting the brown-like adipocyte phenotypes in C3H10T1/2 cells. Moreover, taurine treatment enhanced AMPK phosphorylation in vitro and in vivo, and knockdown of AMPKα1 prevented taurine-mediated induction of PGC1α in C3H10T1/2 cells. Consistently, specific knockdown of PGC1α in iWAT of the HFD-fed mice inhibited taurine-induced browning of iWAT, with the role of taurine in the enhancement of adaptive thermogenesis, the prevention of obesity, and the improvement of insulin sensitivity being partially impaired. These results reveal a functional role of taurine in facilitating the browning of white adipose tissue, which depends on the induction of PGC1α. Our studies also suggest a potential mechanism for the protective role of taurine against obesity, which involves taurine-mediated browning of white adipose tissue.


Subject(s)
Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/pathology , Adipose Tissue, White/drug effects , Adipose Tissue, White/pathology , Obesity/drug therapy , Obesity/pathology , Taurine/pharmacology , AMP-Activated Protein Kinases/metabolism , Adipocytes/drug effects , Adipocytes/pathology , Animals , Anti-Obesity Agents/pharmacology , Anti-Obesity Agents/therapeutic use , Energy Metabolism/drug effects , Gene Expression Regulation/drug effects , Insulin Resistance , Mice , Obesity/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Signal Transduction/drug effects , Taurine/therapeutic use , Thermogenesis/drug effects
9.
J Biol Chem ; 294(31): 11805-11816, 2019 08 02.
Article in English | MEDLINE | ID: mdl-31197036

ABSTRACT

Hepatic steatosis is a hallmark of nonalcoholic fatty liver disease (NAFLD) and is promoted by dysregulated de novo lipogenesis. ATP-citrate lyase (ACLY) is a crucial lipogenic enzyme that is up-regulated in individuals with NAFLD. A previous study has shown that acetylation of ACLY at Lys-540, Lys-546, and Lys-554 (ACLY-3K) increases ACLY protein stability by antagonizing its ubiquitylation, thereby promoting lipid synthesis and cell proliferation in lung cancer cells. But the functional importance of this regulatory mechanism in other cellular or tissue contexts or under other pathophysiological conditions awaits further investigation. Here, we show that ACLY-3K acetylation also promotes ACLY protein stability in AML12 cells, a mouse hepatocyte cell line, and found that the deacetylase sirtuin 2 (SIRT2) deacetylates ACLY-3K and destabilizes ACLY in these cells. Of note, the livers of mice and humans with NAFLD had increased ACLY protein and ACLY-3K acetylation levels and decreased SIRT2 protein levels. Mimicking ACLY-3K acetylation by replacing the three lysines with three glutamines (ACLY-3KQ variant) promoted lipid accumulation both in high glucose-treated AML12 cells and in the livers of high-fat/high-sucrose (HF/HS) diet-fed mice. Moreover, overexpressing SIRT2 in AML12 cells inhibited lipid accumulation, which was more efficiently reversed by overexpressing the ACLY-3KQ variant than by overexpressing WT ACLY. Additionally, hepatic SIRT2 overexpression decreased ACLY-3K acetylation and its protein level and alleviated hepatic steatosis in HF/HS diet-fed mice. Our findings reveal a posttranscriptional mechanism underlying the up-regulation of hepatic ACLY in NAFLD and suggest that the SIRT2/ACLY axis is involved in NAFLD progression.


Subject(s)
ATP Citrate (pro-S)-Lyase/metabolism , Non-alcoholic Fatty Liver Disease/pathology , ATP Citrate (pro-S)-Lyase/antagonists & inhibitors , ATP Citrate (pro-S)-Lyase/genetics , Acetylation , Animals , Cell Line , Diet, High-Fat , Glucose/pharmacology , Humans , Lipid Metabolism/drug effects , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mutagenesis, Site-Directed , Non-alcoholic Fatty Liver Disease/metabolism , Protein Stability , RNA Interference , RNA, Small Interfering/metabolism , Sirtuin 2/genetics , Sirtuin 2/metabolism
10.
J Biol Chem ; 294(24): 9642-9654, 2019 06 14.
Article in English | MEDLINE | ID: mdl-31061100

ABSTRACT

ß-Catenin signaling is triggered by WNT proteins and is an important pathway that negatively regulates adipogenesis. However, the mechanisms controlling the expression of WNT proteins during adipogenesis remain incompletely understood. Lysine demethylase 5A (KDM5A) is a histone demethylase that removes trimethyl (me3) marks from lysine 4 of histone 3 (H3K4) and serves as a general transcriptional corepressor. Here, using the murine 3T3-L1 preadipocyte differentiation model and an array of biochemical approaches, including ChIP, immunoprecipitation, RT-qPCR, and immunoblotting assays, we show that Kdm5a is a target gene of CCAAT/enhancer-binding protein ß (C/EBPß), an important early transcription factor required for adipogenesis. We found that C/EBPß binds to the Kdm5a gene promoter and transactivates its expression. We also found that siRNA-mediated KDM5A down-regulation inhibits 3T3-L1 preadipocyte differentiation. The KDM5A knockdown significantly up-regulates the negative regulator of adipogenesis Wnt6, having increased levels of the H3K4me3 mark on its promoter. We further observed that WNT6 knockdown significantly rescues adipogenesis inhibited by the KDM5A knockdown. Moreover, we noted that C/EBPß negatively regulates Wnt6 expression by binding to the Wnt6 gene promoter and repressing Wnt6 transcription. Further experiments indicated that KDM5A interacts with C/EBPß and that their interaction cooperatively inhibits Wnt6 transcription. Of note, C/EBPß knockdown impaired the recruitment of KDM5A to the Wnt6 promoter, which had higher H3K4me3 levels. Our results suggest a mechanism involving C/EBPß and KDM5A activities that down-regulates the Wnt/ß-catenin pathway during 3T3-L1 preadipocyte differentiation.


Subject(s)
Adipocytes/cytology , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Differentiation , Retinoblastoma-Binding Protein 2/metabolism , Transcriptional Activation , Wnt1 Protein/metabolism , beta Catenin/metabolism , 3T3-L1 Cells , Adipocytes/metabolism , Adipogenesis , Animals , CCAAT-Enhancer-Binding Protein-beta/genetics , Gene Expression Regulation , Histones/genetics , Histones/metabolism , Mice , Promoter Regions, Genetic , Retinoblastoma-Binding Protein 2/genetics , Wnt1 Protein/genetics , beta Catenin/genetics
11.
PLoS One ; 8(6): e65778, 2013.
Article in English | MEDLINE | ID: mdl-23840365

ABSTRACT

BACKGROUND: Myeloperoxidase (MPO) is an endogenous oxidant enzyme that produces reactive oxygen species (ROS) and may be involved in lung carcinogenesis. The MPO-463G>A polymorphism influences MPO transcription and has been associated with lung cancer susceptibility. However, the association between the MPO-463G>A polymorphism and lung cancer risk remains controversial. METHOD: To investigate the effect of this polymorphism on lung cancer susceptibility, we performed a meta-analysis based on 22 published case-control studies including 7,520 patients with lung cancer and 8,600 controls. Odds ratios (ORs) with 95% confidence intervals (CIs) were used to assess the strength of the association. RESULTS: Overall, there was no evidence for significant association between MPO-463G>A polymorphism and lung cancer susceptibility (for AA versus GG: OR = 0.91, 95%CI = 0.67-1.24; for GA versus GG: OR = 0.87, 95% CI = 0.78-0.98; for AA/GA versus GG: OR = 0.90, 95% CI = 0.80-1.01; for AA versus GA/GG: OR = 0.96, 95% CI = 0.72-1.28). In the stratified analyses by ethnicity, source of controls and smoking status, we also did not find any significant association between them. CONCLUSIONS: In summary, this meta-analysis suggests MPO-463G>A polymorphism may not be a risk factor for developing lung cancer. However, further prospective well-designed population-based studies with larger sample size are expected to validate the results.


Subject(s)
Lung Neoplasms/genetics , Peroxidase/genetics , Case-Control Studies , Gene Frequency , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Models, Genetic , Polymorphism, Single Nucleotide , Risk Factors
13.
Zhong Xi Yi Jie He Xue Bao ; 7(5): 463-7, 2009 May.
Article in Chinese | MEDLINE | ID: mdl-19435562

ABSTRACT

OBJECTIVE: To observe sublingual vein characteristics and the expressions of vascular endothelial growth factor (VEGF) and hypoxia-inducible factor 1alpha (HIF-1alpha) proteins in sublingual tissues of Beagle dogs with cirrhotic portal hypertension. METHODS: Twelve Beagle dogs were randomly divided into normal control group and cirrhotic portal hypertension group. There were 6 dogs in each group. A canine model of cirrhosis portal hypertension was established by injecting dimethylnitrosamine (DMN) into portal vein once a week for 7 weeks. The characteristics of sublingual vein were observed. Portal venous pressure was measured by using bioelectric recording techniques. The expressions of VEGF and HIF-1alpha proteins in sublingual vein were detected by immunohistochemical method. RESULTS: The shape and color of sublingual vein in beagle dogs in the cirrhotic portal hypertension group changed obviously as compared with the normal control group. Immunohistochemical results showed that there were almost no expressions of VEGF and HIF-1alpha proteins in sublingual tissues in the normal control group; however, the expressions of VEGF and HIF-1alpha proteins in sublingual tissues in the cirrhotic portal hypertension group significantly increased. CONCLUSION: Changes of portal pressure may lead to the formation of the abnormal sublingual vein by increasing the expressions of VEGF and HIF-1alpha proteins in sublingual tissues in Beagle dogs with portal hypertension.


Subject(s)
Hypertension, Portal/metabolism , Hypertension, Portal/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Tongue/blood supply , Vascular Endothelial Growth Factor A/metabolism , Animals , Dogs , Female , Hypertension, Portal/etiology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Liver Cirrhosis/complications , Male , Medicine, Chinese Traditional , Random Allocation , Tongue/metabolism , Vascular Endothelial Growth Factor A/genetics , Veins/pathology
14.
Acta Crystallogr Sect E Struct Rep Online ; 65(Pt 1): m1, 2008 Dec 03.
Article in English | MEDLINE | ID: mdl-21581467

ABSTRACT

The mol-ecule of the title complex, [Cu(C(6)H(2)N(3)O(7))(2)(C(3)H(7)NO)(2)], is disposed about a crystallographic centre of symmetry. The Cu(II) cation is six-coordinated by two phenolate O atoms and two ortho-nitro O atoms of two picrate units and by two carbonyl O atoms from two coordinated dimethyl-formamide mol-ecules, forming a distorted octa-hedral geometry.

SELECTION OF CITATIONS
SEARCH DETAIL
...