Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Ethnopharmacol ; 324: 117780, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38278377

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Fufang Luohanguo Qingfei granules (LQG) is a Chinese patent medicine, clinically used to treat flu-like symptoms including cough with yellow phlegm, impeded phlegm, dry throat and tongue. However, the protective activity of LQG against influenza infection is indeterminate. AIM OF THE STUDY: This study is to investigate the therapeutic effect of LQG on influenza infection and elucidate its underlying mechanism. MATERIALS AND METHODS: In vivo: A viral susceptible mouse model induced by restraint stress was established to investigate LQG's beneficial effects on influenza susceptibility. MAVS knockout (Mavs-/-) mice were used to verify the potential mechanism of LQG. In vitro: Corticosteroid (CORT)-treated A549 cells were employed to identify the active ingredients in LQG. Mice morbidity and mortality were monitored daily for 21 days. Histopathologic changes and inflammatory cytokines in lung tissues were examined by H&E staining and ELISA. RNA-seq was used to explore the signaling pathway influenced by LQG and further confirmed by qPCR. Immunoblotting and immunohistochemistry (IHC) were used to determine the protein levels. CO-IP and DARTS were applied to detect protein-protein interaction and compound-protein interaction, respectively. RESULTS: LQG effectively attenuated the susceptibility of restrained mice to H1N1 infection. LQG significantly boosted the production of IFN-ß transduced by mitochondrial antiviral-signaling protein (MAVS), while MAVS deficiency abrogated its protective effects on restrained mice infected with H1N1. Moreover, in vitro studies further revealed that mogroside Ⅱ B, amygdalin, and luteolin are potentially active components of LQG. CONCLUSION: These results suggested that LQG inhibited the mitofusin 2 (Mfn2)-mediated ubiquitination of MAVS by impeding the E3 ligase synoviolin 1 (SYVN1) recruitment, thereby enhancing IFN-ß antiviral response. Overall, our work elaborates a potential regimen for influenza treatment through reduction of stress-induced susceptibility.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza, Human , Interferon Type I , Animals , Mice , Humans , Interferon Type I/pharmacology , Interferon Type I/therapeutic use , Influenza, Human/drug therapy , Signal Transduction , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Immunity, Innate
2.
Acta Pharmacol Sin ; 43(7): 1843-1856, 2022 Jul.
Article in English | MEDLINE | ID: mdl-34845369

ABSTRACT

Ras has long been viewed as a promising target for cancer therapy. Farnesylthiosalicylic acid (FTS), as the only Ras inhibitor has ever entered phase II clinical trials, has yielded disappointing results due to its strong hydrophobicity, poor tumor-targeting capacity, and low therapeutic efficiency. Thus, enhancing hydrophilicity and tumor-targeting capacity of FTS for improving its therapeutic efficacy is of great significance. In this study we conjugated FTS with a cancer-targeting small molecule dye IR783 and characterized the anticancer properties of the conjugate FTS-IR783. We showed that IR783 conjugation greatly improved the hydrophilicity, tumor-targeting and therapeutic potential of FTS. After a single oral administration in Balb/c mice, the relative bioavailability of FTS-IR783 was increased by 90.7% compared with FTS. We demonstrated that organic anion transporting polypeptide (OATP) and endocytosis synergistically drove the uptake of the FTS-IR783 conjugate in breast cancer MDA-MB-231 cells, resulting in superior tumor-targeting ability of the conjugate both in vitro and in vivo. We further revealed that FTS-IR783 conjugate could bind with and directly activate AMPK rather than affecting Ras, and subsequently regulate the TSC2/mTOR signaling pathway, thus achieving 2-10-fold increased anti-cancer therapeutic efficacy against 6 human breast cancer cell lines compared to FTS both in vivo and in vitro. Overall, our data highlights a promising approach for the modification of the anti-tumor drug FTS using IR783 and makes it possible to return FTS back to the clinic with a better efficacy.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Farnesol/analogs & derivatives , Farnesol/pharmacology , Farnesol/therapeutic use , Female , Humans , Mice , Salicylates , ras Proteins/metabolism , ras Proteins/therapeutic use
3.
Front Surg ; 9: 932901, 2022.
Article in English | MEDLINE | ID: mdl-36684298

ABSTRACT

Objective: This study aims to explore the application value of computed tomography (CT) three-dimensional (3D) reconstruction, magnetic resonance imaging (MRI) 3D reconstruction, and conventional digital subtraction angiography (DSA) fluoroscopy in percutaneous transhepatic cholangial drainage (PTCD). Methods: The clinical data of 180 patients with obstructive jaundice requiring PTCD from December 2017 to December 2021 were retrospectively analyzed. Following PTCD, CT 3D reconstruction, MRI 3D reconstruction, and conventional DSA fluoroscopy were conducted, after which the surgical success rates, liver function results, and postsurgical complications were compared. Results: The puncture accuracies under CT 3D reconstruction, MRI 3D reconstruction, and conventional DSA fluoroscopy were 90.0% (54/60), 96.7% (58/60), and 80% (48/60), respectively. The degree of jaundice and epigastric discomfort was relieved in all three groups after surgery, while a significant reduction in the levels of total bilirubin and direct bilirubin was observed relative to the levels before surgery (P < 0.05). The incidences of complications in the CT 3D reconstruction, MRI 3D reconstruction, and conventional DSA fluoroscopy groups were 6.7% (4/60), 3.3% (2/60), and 13.3% (8/60), respectively, and the differences among the three groups were statistically significant (P < 0.05). Conclusion: Conducting conventional enhanced CT and MRI scans in patients before surgery might be more practical than the conventional puncture method. Among the methods under study, MRI 3D reconstruction was found to be safer and more feasible than CT 3D reconstruction and conventional DSA fluoroscopy in PTCD. MRI 3D reconstruction could reduce the degree of jaundice, improve the success rate of surgery, reduce the incidence of complications due to surgery, and improve the patients' tolerance to surgery.

4.
Int Arch Allergy Immunol ; 182(3): 210-219, 2021.
Article in English | MEDLINE | ID: mdl-33477144

ABSTRACT

BACKGROUND: This study aims to investigate the regulatory effect of microRNA-96-5p (miR-96-5p) in the pathophysiological process of allergic rhinitis (AR). METHODS: Nasal mucosal tissue samples were collected from AR patients and healthy controls. An in vitro AR model was established by stimulating human nasal epithelial cells (HNECs) with interleukin (IL)-13. The expressions of target genes and proteins were measured by qPCR, Western blot, or ELISA. Dual-luciferase reporter assay and pull-down assay were performed to confirm the interaction between miR-96-5p and DEP domain-containing mammalian target of rapamycin-interacting protein (DEPTOR). RESULTS: The level of miR-96-5p was increased while the expression of DEPTOR was decreased in AR patients. The expressions of proinflammatory cytokines were markedly increased and the mammalian target of rapamycin (mTOR)/NF-κB pathway was activated in HNECs following IL-13 stimulation. miR-96-5p downregulation alleviated the stimulated function by IL-13. DEPTOR was the target of miR-96-5p. Knockdown of DEPTOR reversed the function of miR-96-5p inhibitor on IL-13-stimulated HNECs. CONCLUSIONS: The current study showed that miR-96-5p and DEPTOR were aberrantly expressed in AR nasal mucosa. miR-96-5p knockdown inhibited the production of inflammatory cytokines and the activation of mTOR/NF-κB pathway via targeting DEPTOR. These findings suggested that miR-96-5p might be used as a diagnostic marker and therapeutic target for the treatment of AR.


Subject(s)
Gene Expression Regulation , Intracellular Signaling Peptides and Proteins/metabolism , MicroRNAs/genetics , NF-kappa B/metabolism , Rhinitis, Allergic/etiology , Rhinitis, Allergic/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Cytokines/genetics , Cytokines/metabolism , Humans , Inflammation Mediators/metabolism , Nasal Mucosa/immunology , Nasal Mucosa/metabolism , Nasal Mucosa/pathology , RNA Interference , Rhinitis, Allergic/diagnosis
5.
Am J Transl Res ; 12(10): 6975, 2020.
Article in English | MEDLINE | ID: mdl-33194088

ABSTRACT

[This corrects the article on p. 2570 in vol. 12, PMID: 32655791.].

6.
Am J Transl Res ; 12(6): 2570-2584, 2020.
Article in English | MEDLINE | ID: mdl-32655791

ABSTRACT

An impaired epithelial barrier is often observed in allergic rhinitis (AR), which facilitates the infiltration of allergens. The aim of this study was to investigate the role of autophagy in the impaired epithelial barrier in AR and the related signalling pathways. A human nasal epithelial cell line was treated with dust mite allergen (Derp1). Autophagy was evaluated by GFP-LC3 adenovirus transfection and measurement of autophagy-related proteins. Epithelial barrier function was evaluated by measuring tight junction protein expression, transepithelial electrical resistance, and fluorescein isothiocyanate-dextran (FD4) permeability. Next, miR-125b inhibitor, miR-125b mimics, shRNA targeting FoxP3, pcDNA3.1 expressing FoxP3, and inhibitor of C-X-C motif chemokine receptor type 4 (CXCR4) were used to investigate the roles of miR-125b, FoxP3, and CXCR4 in epithelial cell autophagy and epithelial barrier function. An in vivo AR model was generated by exposing rat nasal mucosa to an allergen. Derp1 exposure enhanced autophagy and impaired the epithelial barrier in epithelial cells. Upregulation of miR-125b expression led to enhanced autophagy and impaired epithelial barrier through inhibition of FoxP3. Derp1 exposure increased miR-125b expression by increasing the expression and activation of CXCR4, which downregulated FoxP3 expression and led to enhanced autophagy and an impaired epithelial barrier. In vivo analysis confirmed the role of the CXCR4/miR-125b/FoxP3 axis in the impaired epithelial barrier in AR. This study demonstrates that the CXCR4/miR-125b/FoxP3 axis may participate in the pathogenesis of AR by regulating autophagy in epithelial cells and dysfunction of the epithelial barrier.

7.
Cell Cycle ; 19(10): 1132-1142, 2020 05.
Article in English | MEDLINE | ID: mdl-32298206

ABSTRACT

Nasal epithelial cells are the first barrier against allergen infiltration in allergic rhinitis (AR), and the relationship between nasal epithelial cells and mast cell-mediated hypersensitivity remains unclear. This study aimed to investigate the possible association between allergen-challenged nasal epithelial cells (AR-HNEpC) and mast cell degranulation in AR. Our data revealed that calcium influx and degranulation were increased in AR-HNEpC-co-cultured mast cells. Expression of IL-33, a factor that binds to ST2 receptors on mast cells and regulates their degranulation, was elevated in AR-HNEpC. Blocking IL-33/ST2 pathway activated autophagy and inhibited degranulation and inflammatory factor release in mast cells. Furthermore, PI3K/mTOR was increased in IL-33-treated mast cells. Inhibition on PI3K/mTOR pathway enhanced autophagy and inhibited degranulation. Analysis using an in vivo AR model supported the above findings. In conclusion, IL-33 from epithelial cells promotes degranulation of mast cells in AR through inhibition on ST2/PI3K/mTOR-mediated autophagy, which provides a potential therapeutic target for the disease.Abbreviations: AR: allergic rhinitis; IL: interleukin; TNF-α: tumor necrosis factor-alpha; INF-γ: interferon-gamma; HNEpC: human nasal epithelial cell line; ATCC: American Type Culture Collection; C48/80: compound 48/80; 3-MA: 3-methyladenine; qPCR: quantitative PCR; AR-HNEpC: dust mite allergen-treated nasal epithelial cells; IgE: immunoglobulin E; Atg7: autophagy-related gene 7.


Subject(s)
Autophagy/genetics , Cell Degranulation/genetics , Epithelial Cells/metabolism , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-33/metabolism , Mast Cells/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Rhinitis, Allergic/metabolism , TOR Serine-Threonine Kinases/metabolism , Allergens/pharmacology , Animals , Cell Line , Disease Models, Animal , Epithelial Cells/drug effects , Humans , Interleukin-33/genetics , Pyroglyphidae/immunology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Signal Transduction/genetics , Transfection
8.
Yi Chuan ; 42(1): 112-125, 2020 Jan 20.
Article in English | MEDLINE | ID: mdl-31956101

ABSTRACT

Alcohol abuse causes tissue and organ damage, and may participate neuropsychiatric diseases. Studies have shown that DNA methylation plays an important role in gene expression and behavioral changes induced by alcohol, however the causative neurobiological mechanisms have not been clarified. In this study, 32 healthy adult male SD rats were randomly divided into a drinking water control group (n=16) and a chronic alcohol exposure group (n=16). The alcohol preference and locomotor activity of rats were evaluated by two-bottle choice test (TBCT) and open-field test (OFT). DNA methylation in the medial prefrontal cortex (mPFC) tissue was detected by the reduced representative bisulfite sequencing (RRBS) technology. The methylation differential genes closely related to alcohol abuse were screened. qRT-PCR was used to verify the mRNA expression patterns of differential genes. qRT-PCR and Western blot were used to detect the expression of DNA methyltransferases (DNMTs) and methyl CpG binding protein 2 (MeCP2). Furthermore, the effect of short-term alcohol exposure (7 days) on DNMTs and MeCP2 in the mPFC of rats was tested (n=8/group). The results indicated that the methylation level of promoter region in the mPFC of rats exposed to chronic alcohol was significantly increased. In addition, the increased methylation levels in the promoter of Ntf3 and Ppm1G were accompanied by down-regulated mRNA levels in the chronic alcohol exposure group. The decreased methylation levels in the promoter of Hap1 and DUSP1 were accompanied by up-regulated mRNA levels. Furthermore, chronic alcohol exposure increased the mRNA and protein levels of DNMT3B and MeCP2. However, short term alcohol exposure did not affect their expression. This present study provides evidence that DNA methylation is associated with the development of alcohol abuse, which may be regulated by DNMT3B and MeCP2. The target genes Ntf3, Ppm1G, Hap1, and DUSP1 related to alcohol abuse were discovered as well, providing new insights into the neurobiological mechanism of alcohol abuse and the potential pharmacological targets for the treatment of alcohol abuse.


Subject(s)
Alcoholism/physiopathology , Behavior, Animal , DNA Methylation , Prefrontal Cortex/physiopathology , Animals , DNA (Cytosine-5-)-Methyltransferases/genetics , Locomotion , Male , Methyl-CpG-Binding Protein 2/genetics , Promoter Regions, Genetic , Rats , Rats, Sprague-Dawley , DNA Methyltransferase 3B
9.
Front Pharmacol ; 9: 434, 2018.
Article in English | MEDLINE | ID: mdl-29765324

ABSTRACT

Lung cancer is a leading cause of cancer-related deaths worldwide. NOTCH3 signaling is mainly expressed in non-small cell lung carcinoma (NSCLC), and has been proposed as a therapeutic target of NSCLC. While, few agents for preventing or treating NSCLC via targeting NOTCH3 signaling are used in modern clinical practice. Evodiamine (EVO), an alkaloid derived from Euodiae Fructus, possesses low toxicity and has long been shown to exert anti-lung cancer activity. However, the underlying anti-lung cancer mechanisms of EVO are not yet fully understood. In this study, we explored the involvement of NOTCH3 signaling in the anti-lung cancer effects of EVO. Urethane-induced lung cancer mouse model and two NSCLC cell models, A549 and H1299, were used to evaluate the in vivo and in vitro anti-lung cancer action of EVO. A DNA methyltransferase inhibitor was employed to investigate the role of NOTCH3 signaling in the anti-lung cancer effects of EVO. Results showed that EVO potently reduced tumor size and tumor numbers in mice, and inhibited NOTCH3 in the tumors. EVO also dramatically reduced cell viability, induced G2/M cell cycle arrest, inhibited cell migration and reduced stemness in cultured NSCLC cells. Mechanistic studies showed that EVO potently inhibited NOTCH3 signaling by activation of DNMTs-induced NOTCH3 methylation. Importantly, inhibition of NOTCH3 methylation in NSCLC cells diminished EVO's anti-NSCLC effects. Collectively, EVO, a novel NOTCH3 methylation stimulator, exerted potent anti-lung cancer effects partially by inhibiting NOTCH3 signaling. These findings provide new insight into the EVO's anti-NSCLC action, and suggest a potential role of EVO in lung cancer prevention and treatment.

10.
Bioorg Med Chem Lett ; 20(3): 805-8, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20064722

ABSTRACT

An efficient total synthesis of (R) and (S)-3-methyl 5-pentyl 2,6-dimethyl-4-(3-nitrophenyl)-1,4-dihydropyridine-3,5-dicarboxylate in high optical purities is reported. The useful step is the resolution of racemic 2, 6-dimethyl-5-methoxycarbonyl-4-(3-nitrophenyl)-1,4-dihydropyridine-3-carboxylic acid by using commercially available Cinchona alkaloids cinchonidine and quinidine as the resolving agents. Under the optimum conditions, the optical purities for R- and S-enantiomers are extremely high (ee>99.5%). The further dihydropyridine receptor binding activity assay shows that the S-enantiomer is more potent than R-enantiomer both in rat cardiac (approximately 19 times) and cerebral cortex membrane (12 times).


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium/metabolism , Dicarboxylic Acids/chemical synthesis , Dicarboxylic Acids/metabolism , Dihydropyridines/chemical synthesis , Dihydropyridines/metabolism , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Dicarboxylic Acids/pharmacology , Dihydropyridines/pharmacology , Myocardium/metabolism , Protein Binding/drug effects , Protein Binding/physiology , Rats , Stereoisomerism
11.
Can J Physiol Pharmacol ; 87(1): 51-5, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19142215

ABSTRACT

Approximately 50% of hypertensive patients are postmenopausal women; therefore, any antihypertensive therapy must not adversely affect bone loss in this population. Recently, however, concern has been raised that use of angiotensin AT1 receptor antagonists may increase the tendency to develop postmenopausal osteoporosis by decreasing transforming growth factor-beta1 (TGF-beta1), which has been implicated in bone mass maintenance. In the present study, we selected telmisartan and valsartan as representatives of angiotensin AT1 receptor antagonists and used ovariectomized (OVX) rats as a model of human postmenopausal osteoporosis. After 3 months treatment with telmisartan (5 mg/kg daily) or valsartan (10 mg/kg daily), OVX rats showed no signs of adverse effects on bone mineral density of the lumbar vertebrae (L1-L5) or the total femur, nor did treatment affect serum levels of osteocalcin and osteoclast-derived tartrate-resistant acid phosphatase (TRACP-5b). Bone TGF-beta1 content remained unchanged, although treatment with telmisartan and valsartan significantly reduced serum TGF-beta1 levels (p < 0.05). In conclusion, chronic treatment with angiotensin AT1 receptor antagonists reduced serum but not bone TGF-beta1 levels and did not accelerate ovariectomy-induced bone loss in rats.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/toxicity , Bone and Bones/drug effects , Osteoporosis, Postmenopausal/chemically induced , Transforming Growth Factor beta1/analysis , Acid Phosphatase/blood , Animals , Benzimidazoles/toxicity , Benzoates/toxicity , Bone Density/drug effects , Bone and Bones/chemistry , Female , Humans , Isoenzymes/blood , Osteocalcin/blood , Osteoporosis, Postmenopausal/metabolism , Ovariectomy , Rats , Rats, Sprague-Dawley , Tartrate-Resistant Acid Phosphatase , Telmisartan , Tetrazoles/toxicity , Transforming Growth Factor beta1/blood , Valine/analogs & derivatives , Valine/toxicity , Valsartan
12.
Phytother Res ; 22(3): 389-94, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18058992

ABSTRACT

The purpose of the present study was to investigate the effects of the Chinese medical herb Astragali Radix on myocardial injury in vivo and its possible mechanisms. Myocardial injury in rats was induced by the subcutaneous injection of a high dose of isoproterenol for 10 days, and the therapeutic effects of Astragali Radix were observed. Cardiac hemodynamics, heart coefficient and marker enzymes in serum showed that Astragali Radix prevented isoproterenol-induced myocardial damage. Astragali Radix also improved the antioxidant status by decreasing the lipid peroxidative product malondialdehyde and increasing the activity of the antioxidant enzyme superoxide dismutase. The observed depressions in sarcoplasmic reticulum Ca2+-ATPase mRNA and protein expression as well as Ser(16)-phosphorylated phospholamban protein expression in isoproterenol-treated rats were attenuated by Astragali Radix treatment. Moreover, treatment with Astragali Radix showed higher myocardial cAMP content compared with the isoproterenol-alone group. These results suggest that the antioxidant property and partial prevention of changes in protein and gene expression of cardiac sarcoplasmic reticulum Ca2+ regulatory proteins which may be mediated through the cAMP pathway could help to explain the beneficial effects of Astragali Radix on myocardial injury in vivo.


Subject(s)
Astragalus Plant/chemistry , Cardiomyopathies/drug therapy , Cardiotonic Agents/pharmacology , Drugs, Chinese Herbal/pharmacology , Heart/drug effects , Animals , Cardiomegaly/drug therapy , Cardiomyopathies/chemically induced , Carrier Proteins/analysis , Carrier Proteins/biosynthesis , Cyclic AMP/analysis , Enzymes/analysis , Enzymes/drug effects , Gene Expression/drug effects , Hemodynamics/drug effects , Isoproterenol/pharmacology , Male , Malondialdehyde/analysis , Models, Animal , Myocardium/pathology , Rats , Rats, Sprague-Dawley
13.
Eur J Pharmacol ; 568(1-3): 203-12, 2007 Jul 30.
Article in English | MEDLINE | ID: mdl-17509559

ABSTRACT

Astragaloside IV, the primary pure saponin isolated from Astragalus membranaceus has been found to have potent cardioprotective effects. In this study, we aim to investigate if the beneficial effects of astragaloside IV on cardiac function are associated with improvement in sarcoplasmic reticulum Ca(2+)-pump function in myocardial injury in vivo. Myocardial injury in rats was induced by subcutaneous injection of a high dose of isoproterenol, and the therapeutic effect of astragaloside IV was observed. Isoproterenol-treated rats showed widespread subendocardial necrosis, a rise in serum lactate dehydrogenase and creatine kinase, formation of lipid oxide product malondialdehyde and inhibition of left ventricular diastolic and systolic function, which suggested severe myocardial injury and acute heart failure. Moreover, sarcoplasmic reticulum Ca(2+)-uptake ability and Ca(2+)-ATPase (SERCA2a) activity were significantly reduced. And the level of SERCA2a mRNA and protein expression was also markedly decreased, associated with a decrease in Ser(16)-phosphorylated phospholamban protein expression, while total phospholamban level was unchanged in the isoproterenol-treated group compared with controls. However, these biochemical and hemodynamic changes in the acute failing hearts were prevented by treatment of isoproterenol-induced rats with astragaloside IV. Likewise, the observed reductions in sarcoplasmic reticulum Ca(2+)-pump function as well as in SERCA2a mRNA and protein levels and the phosphorylation level of phospholamban in the injured hearts were attenuated by astragaloside IV treatment. These results suggest that the beneficial effect of astragaloside IV on isoproterenol-induced myocardial injury may be due to its ability to prevent changes of SERCA2a and Ser(16)-phosphorylated phospholamban protein expression and, thus, may prevent the depression in sarcoplasmic reticulum Ca(2+) transport and improve cardiac function.


Subject(s)
Drugs, Chinese Herbal/therapeutic use , Heart Injuries/drug therapy , Heart/drug effects , Saponins/therapeutic use , Sarcoplasmic Reticulum/drug effects , Triterpenes/therapeutic use , Animals , Blood Pressure/drug effects , Calcium/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Calcium-Transporting ATPases/metabolism , Creatine Kinase/blood , Heart/physiology , Heart Injuries/metabolism , Heart Injuries/pathology , Heart Injuries/physiopathology , Heart Rate/drug effects , L-Lactate Dehydrogenase/blood , Male , Malondialdehyde/metabolism , Myocardium/metabolism , Myocardium/pathology , Phosphorylation , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Superoxide Dismutase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...