Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Article in English | MEDLINE | ID: mdl-38767671

ABSTRACT

Activation of the renin-angiotensin system (RAS) triggers oxidative stress and an inflammatory response in the hypothalamic paraventricular nucleus (PVN), in turn increasing the sympathetic hyperactivity that is a major cause of hypertension. Pyridostigmine has cardioprotective effects by suppressing the RAS of myocardial tissue. However, whether pyridostigmine attenuates hypertension by inhibiting the RAS of the PVN remains unclear. We thus investigated the effect and mechanism of pyridostigmine on two-kidney one-clip (2K1C)-induced hypertension. 2K1C rats received pyridostigmine, or not, for 8 weeks. Cardiovascular function, hemodynamic parameters, and autonomic activity were measured. The PVN levels of pro-/anti-inflammatory cytokines, oxidative stress, and RAS signaling molecules were evaluated. Our results showed that hypertension was accompanied by cardiovascular dysfunction and an autonomic imbalance characterized by enhanced sympathetic but diminished vagal activity. The PVN levels of interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), reactive oxygen species (ROS), NOX-2, and malondialdehyde (MDA) increased; those of IL-10 and superoxide dismutase (SOD) decreased. Moreover, the RAS signaling pathway was activated, as evidenced by increased levels of the angiotensin-converting enzyme (ACE), angiotensin II (Ang II), and the Ang II type 1 receptor (AT1R) and a decreased AT2R level. Pyridostigmine lowered blood pressure and improved cardiovascular function, associated with restoration of the autonomic balance. Meanwhile, pyridostigmine decreased PVN IL-6, TNF-α, ROS, NOX-2, and MDA levels and increased IL-10 and SOD levels. Additionally, pyridostigmine suppressed PVN ACE, Ang II, and AT1R levels and increased AT2R expression. Pyridostigmine attenuated hypertension by inhibiting PVN oxidative stress and inflammation induced by the RAS.

2.
Front Pharmacol ; 13: 923395, 2022.
Article in English | MEDLINE | ID: mdl-35860018

ABSTRACT

Background: Shufeng Jiedu (SFJD) capsules can be used as adjunctive treatment for patients with community-acquired pneumonia, but the effectiveness and safety of SFJD are not clear. This review aims to evaluate the effectiveness and safety of SFJD based on randomized controlled trials (RCTs). Methods: A systematic review was conducted by searching PubMed, Embase, Scopus, Web of Science, CENTRAL, CNKI, VIP, CBM, Wanfang and trial registry platforms from their inception to March 2022. Two reviewers screened studies, extracted the data and assessed risk of bias independently. The data were pooled for meta-analysis or presented narratively. Results: Seventeen RCTs involving 1840 participants were included. All trials compared SFJD plus antibiotics to antibiotics, or combined with symptomatic treatment in both groups. The overall certainty of evidence was assessed as moderate to very low certainty. Compared with routine treatment (antibiotics alone or antibiotics plus symptomatic treatment), SFJD plus routine treatment showed beneficial effects in resolution of fever (MD -1.20 days, 95%CI -1.73 to -0.67; 10 RCTs; very low certainty), cough (MD -1.02 days, 95%CI -1.23 to -0.81; 9 RCTs; moderate certainty), phlegm (MD -1.46 days, 95%CI -2.84 to -0.08; 6 RCTs; very low certainty), pulmonary crepitations (MD -1.61 days, 95%CI -2.64 to -0.59; 8 RCTs; low certainty), shortness of breath (MD -2.80 days, 95%CI -2.88 to -2.72; 2 RCTs; low certainty) and chest pain (MD -2.85 days, 95%CI -3.01 to -2.69; 1 RCT; low certainty). There was no significant difference in pathogen clearance (1 RCT). No serious adverse events were reported, but 2.60% (5/192) patients reported nausea in the SFJD groups, 1.04% (2/192) participants in routine group, and no significant difference was identified. Conclusions: Current evidence suggests that adding SFJD may shorten the duration of symptom relief in community-acquired pneumonia for 1-2 days. The adverse events were minor and controllable, and no serious adverse events were reported. Well-reported trials and potential of reducing antibiotics were expected in the future studies.

3.
Antioxidants (Basel) ; 11(2)2022 Jan 31.
Article in English | MEDLINE | ID: mdl-35204171

ABSTRACT

BACKGROUND: Numerous studies have indicated that a high salt diet inhibits brain Na+/K+-ATPase (NKA) activity, and affects oxidative stress and inflammation in the paraventricular nucleus (PVN). Furthermore, Na+/K+-ATPase alpha 2-isoform (NKA α2) may be a target in the brain, taking part in the development of salt-dependent hypertension. Therefore, we hypothesized that NKA α2 regulates oxidative stress and inflammation in the PVN in the context of salt-induced hypertension. METHODS: Part I: We assessed NKA subunits (NKA α1, NKA α2, and NKA α3), Na+/K+-ATPase activity, oxidative stress, and inflammation in a high salt group (8% NaCl) and normal salt group (0.3% NaCl). Part II: NKA α2 short hairpin RNA (shRNA) was bilaterally microinjected into the PVN of salt-induced hypertensive rats to knockdown NKA α2, and we explored whether NKA α2 regulates downstream signaling pathways related to protein kinase C γ (PKC γ)-dependent oxidative stress and toll-like receptor 4 (TLR4)-induced inflammation in the PVN to promote the development of hypertension. RESULTS: High salt diet increased NKA α1 and NKA α2 protein expression in the PVN but had no effect on NKA α3 compared to the normal salt diet. Na+/K+-ATPase activity and ADP/ATP ratio was lower, but NAD(P)H activity and NF-κB activity in the PVN were higher after a high salt diet. Bilateral PVN microinjection of NKA α2 shRNA not only improved Na+/K+-ATPase activity and ADP/ATP ratio but also suppressed PKC γ-dependent oxidative stress and TLR4-dependent inflammation in the PVN, thus decreasing sympathetic activity in rats with salt-induced hypertension. CONCLUSIONS: NKA α2 in the PVN elicits PKC γ/Rac1/NAD (P)H-dependent oxidative stress and TLR4/MyD88/NF-κB-induced inflammation in the PVN, thus increasing MAP and sympathetic activity during the development of salt-induced hypertension.

4.
Toxicol Rep ; 8: 657-667, 2021.
Article in English | MEDLINE | ID: mdl-33868952

ABSTRACT

Oxidative stress is considered to be one of main pathophysiological mechanisms in myocardial ischemia/reperfusion (I/R) injury. Lycium barbarum polysaccharides (LBP), the main ingredient of Lycium barbarum, have potential antioxidant activity. We aimed to investigate the effects of LBP on myocardial I/R injury and explore the underlying mechanisms. Myocardial I/R group was treated with or without LBP to evaluate oxidative stress markers and the role of Nrf2 signal pathway. Our results showed that I/R increased infarct size and the activities of creatine kinase (CK) and lactate dehydrogenase (LDH) when compared with control group. Meanwhile, the levels of reactive oxygen species (ROS), malondialdehyde (MDA), interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) were enhanced and the activities of superoxide dismutase (SOD), glutathione peroxidase (GPX) and catalase (CAT) were decreased. These changes were associated with a significant increase in myocardial apoptosis, ultimately leading to cardiac dysfunction. LBP reduced infarct size (38.4 ± 2 % versus 19.4 ± 1.8 %, p < 0.05), CK and LDH activities and myocardial apoptotic index. Meanwhile, LBP suppressed the production of ROS and restored redox status. Additionally, LBP increased protein level of nuclear Nrf2 in vivo (2.1 ± 0.3 versus 3.8 ± 0.4, p < 0.05) and in vitro (1.9 ± 0.2 versus 3.8 ± 0.1, p < 0.05) and subsequently upregulated heme oxygenase 1 and NADPH dehydrogenase quinone 1 compared to I/R group. Interestingly, Nrf2 siRNA abolished the protective effects of LBP. LBP suppressed oxidative stress damage and attenuated cardiac dysfunction induced by I/R via activation of the Nrf2 antioxidant signal pathway.

5.
J Cardiovasc Pharmacol ; 77(2): 170-181, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33538532

ABSTRACT

ABSTRACT: Oxidative stress, the renin-angiotensin system (RAS), and inflammation are some of the mechanisms involved in the pathogenesis of hypertension. The aim of this study is to examine the protective effect of the chronic administration of astaxanthin, which is extracted from the shell of crabs and shrimps, into hypothalamic paraventricular nucleus (PVN) in spontaneously hypertensive rats. Animals were randomly assigned to 2 groups and treated with bilateral PVN infusion of astaxanthin or vehicle (artificial cerebrospinal fluid) through osmotic minipumps (Alzet Osmotic Pumps, Model 2004, 0.25 µL/h) for 4 weeks. Spontaneously hypertensive rats had higher mean arterial pressure and plasma level of norepinephrine and proinflammatory cytokine; higher PVN levels of reactive oxygen species, NOX2, NOX4, IL-1ß, IL-6, ACE, and AT1-R; and lower PVN levels of IL-10 and Cu/Zn SOD, Mn SOD, ACE2, and Mas receptors than Wistar-Kyoto rats. Our data showed that chronic administration of astaxanthin into PVN attenuated the overexpression of reactive oxygen species, NOX2, NOX4, inflammatory cytokines, and components of RAS within the PVN and suppressed hypertension. The present results revealed that astaxanthin played a role in the brain. Our findings demonstrated that astaxanthin had protective effect on hypertension by improving the balance between inflammatory cytokines and components of RAS.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Antihypertensive Agents/administration & dosage , Arterial Pressure/drug effects , Cytokines/metabolism , Hypertension/drug therapy , Inflammation Mediators/metabolism , Paraventricular Hypothalamic Nucleus/drug effects , Renin-Angiotensin System/drug effects , Animals , Disease Models, Animal , Hypertension/metabolism , Hypertension/physiopathology , Infusions, Parenteral , Male , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/physiopathology , Rats, Inbred SHR , Rats, Inbred WKY , Time Factors , Xanthophylls/administration & dosage
7.
Cardiovasc Toxicol ; 21(4): 286-300, 2021 04.
Article in English | MEDLINE | ID: mdl-33165770

ABSTRACT

Hypertension, as one of the major risk factors for cardiovascular disease, significantly affects human health. Prostaglandin E2 (PGE2) and the E3-class prostanoid (EP3) receptor have previously been demonstrated to modulate blood pressure and hemodynamics in various animal models of hypertension. The PGE2-evoked pressor and biochemical responses can be blocked with the EP3 receptor antagonist, L-798106 (N-[(5-bromo-2methoxyphenyl)sulfonyl]-3-[2-(2-naphthalenylmethyl) phenyl]-2-propenamide). In the hypothalamic paraventricular nucleus (PVN), sympathetic excitation can be introduced by PGE2, which can activate EP3 receptors located in the PVN. In such a case, the central knockdown of EP3 receptor can be considered as a potential therapeutic modality for hypertension management. The present study examined the efficacy of the PVN infusion of L-798106, by performing experiments on spontaneously hypertensive rats (SHRs) and normotensive Wistar-Kyoto rats (WKYs). The rats were administered with chronic bilateral PVN infusion of L-798106 (10 µg/day) or the vehicle for 28 days. The results indicated that the SHRs had a higher mean arterial pressure (MAP), an increased Fra-like (Fra-LI) activity in the PVN, as well as a higher expression of gp91phox, mitogen-activated protein kinase (MAPK), and proinflammatory cytokines in the PVN compared with the WKYs. Additionally, the expression of Cu/Zn-SOD in the PVN of the SHRs was reduced compared with the WKYs. The bilateral PVN infusion of L-798106 significantly reduced MAP, as well as plasma norepinephrine (NE) levels in the SHRs. It also inhibited Fra-LI activity and reduced the expression of gp91phox, proinflammatory cytokines, and MAPK, whereas it increased the expression of Cu/Zn-SOD in the PVN of SHRs. In addition, L-798106 restored the balance of the neurotransmitters in the PVN. On the whole, the findings of the present study demonstrate that the PVN blockade of EP3 receptor can ameliorate hypertension and cardiac hypertrophy partially by attenuating ROS and proinflammatory cytokines, and modulating neurotransmitters in the PVN.


Subject(s)
Antihypertensive Agents/pharmacology , Blood Pressure/drug effects , Hypertension/prevention & control , Inflammation Mediators/metabolism , Oxidative Stress/drug effects , Paraventricular Hypothalamic Nucleus/drug effects , Prostaglandin Antagonists/pharmacology , Receptors, Prostaglandin E, EP3 Subtype/antagonists & inhibitors , Sulfonamides/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Cardiomegaly/metabolism , Cardiomegaly/physiopathology , Cardiomegaly/prevention & control , Disease Models, Animal , Hypertension/metabolism , Hypertension/physiopathology , Male , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/physiopathology , Rats, Inbred SHR , Rats, Inbred WKY , Reactive Oxygen Species/metabolism , Receptors, Prostaglandin E, EP3 Subtype/metabolism , Signal Transduction
8.
J Biochem Mol Toxicol ; 34(11): e22573, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32659049

ABSTRACT

Docosahexaenoic acid (DHA) is reported to have the potential to ameliorate pulmonary arterial hypertension (PAH), while the specific mechanism is still obscure. This study aims to investigate the function of DHA in pulmonary artery smooth muscle cells (PASMCs) and explore the underlying mechanism. In our study, DHA was used to incubate PASMCs. Cytosolic-free Ca2+ concentration ([Ca2+ ]cyt) was measured using Fluo-3 AM method. Real-time polymerase chain reaction was used to detect microRNA-16 (miR-16) and calcium-sensing receptor (CaSR) messenger RNA expression levels. CCK-8 assay, BrdU assay, and Transwell assay were employed to detect the effects of DHA on proliferation and migration of PASMCs. CaSR was confirmed as a direct target of miR-16 using dual-luciferase assay, polymerase chain reaction, and Western blot analysis. It was found that DHA significantly inhibited PASMC proliferation and migration and decreased [Ca2+ ]cyt. After transfection of miR-16 mimics, proliferation and migration ability of PASMCs were significantly inhibited, whereas opposite effects were observed after miR-16 inhibition. [Ca2+ ]cyt was also inhibited by miR-16 transfection. DHA then promoted the expression of miR-16, and the effects of DHA on PASMCs were annulled when miR-16 was inhibited. CaSR was identified as a direct target of miR-16. CaSR was inhibited directly by miR-16 and indirectly by DHA. In conclusion, DHA inhibits the proliferation and migration of PASMCs, and probably ameliorates PAH via regulating miR-16/CaSR axis.


Subject(s)
Calcium/metabolism , Down-Regulation/drug effects , MicroRNAs/metabolism , Muscle, Smooth/drug effects , Pulmonary Artery/drug effects , Receptors, Calcium-Sensing/metabolism , Binding Sites , Cells, Cultured , Docosahexaenoic Acids/pharmacology , Humans , Ion Transport , Muscle, Smooth/cytology , Muscle, Smooth/metabolism , Pulmonary Artery/cytology , Pulmonary Artery/metabolism
9.
Toxicol Appl Pharmacol ; 394: 114953, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32165127

ABSTRACT

Exercise training is one of the major non-pharmacological treatments for hypertension. However, the central mechanism by which exercise training attenuates the hypertensive responses remains unclear. Irisin is a muscle-secreted cytokine derived from fibronectin type III domain containing 5 (FNDC5) that will be released into the circulation during exercise. We hypothesized that irisin may play a role in the blood pressure regulation by exercise. To examine the hypothesis, our study investigated the effect of irisin on hypertension and its central mechanism. The study was performed in spontaneously hypertensive rats (SHRs) and normotensive Wistar-Kyoto rats. We found that intravenous injection of irisin effectively reduced blood pressure, plasma norepinephrine, paraventricular nucleus (PVN) levels of neuronal activation, oxidative stress and inflammation in SHRs. Moreover, irisin activated nuclear factor E2-related factor-2 (Nrf2) and restored the imbalance of neurotransmitters in the PVN. Our study also found PVN knockdown of Nrf2 abolished the protective effects of irisin on hypertension. These findings demonstrate irisin can improve hypertension via Nrf2-mediated antioxidant in the PVN.


Subject(s)
Antihypertensive Agents/pharmacology , Fibronectins/pharmacology , NF-E2-Related Factor 2/drug effects , Paraventricular Hypothalamic Nucleus/drug effects , Signal Transduction/drug effects , Animals , Antioxidants/metabolism , Cytokines/metabolism , Neurotransmitter Agents/metabolism , Norepinephrine/blood , Oxidative Stress/drug effects , Physical Exertion , Rats , Rats, Inbred SHR , Rats, Inbred WKY
10.
J Biochem Mol Toxicol ; 34(5): e22467, 2020 May.
Article in English | MEDLINE | ID: mdl-32040235

ABSTRACT

Oxidative stress induced by long-term cyclosporine A (CsA) administration is a major cause of chronic nephrotoxicity, which is characterized by tubular atrophy, tubular cell apoptosis, and interstitial fibrosis in the progression of organ transplantation. Although hydrogen-rich water (HRW) has been used to prevent various oxidative stress-related diseases, its underlying mechanisms remain unclear. This study investigated the effects of HRW on CsA-induced nephrotoxicity and its potential mechanisms. After administration of CsA (25 mg/kg/day), rats were treated with or without HRW (12 mL/kg) for 4 weeks. Renal function and vascular activity were investigated. Histological changes in kidney tissues were analyzed using Masson's trichrome and terminal deoxynucleotidyl transferase dUTP nick-end labeling stains. Oxidative stress markers and the activation of the Kelch-like ECH-associated protein 1 (Keap1)/nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway were also measured. We found that CsA increased the levels of reactive oxygen species (ROS) and malonaldehyde (MDA), but it reduced glutathione (GSH) and superoxide dismutase (SOD) levels. Such alterations induced vascular dysfunction, tubular atrophy, interstitial fibrosis, and tubular apoptosis. This was evident secondary to an increase in urinary protein, serum creatinine, and blood urea nitrogen, ultimately leading to renal dysfunction. Conversely, HRW decreased levels of ROS and MDA while increasing the activity of GSH and SOD. This was accompanied by an improvement in vascular and renal function. Moreover, HRW significantly decreased the level of Keap1 and increased the expression of Nrf2, NADPH dehydrogenase quinone 1, and heme oxygenase 1. In conclusion, HRW restored the balance of redox status, suppressed oxidative stress damage, and improved kidney function induced by CsA via activation of the Keap1/Nrf2 signaling pathway.


Subject(s)
Cyclosporine/adverse effects , Hydrogen/pharmacology , Immunosuppressive Agents/adverse effects , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/metabolism , Protective Agents/pharmacology , Renal Insufficiency/chemically induced , Signal Transduction/drug effects , Water/pharmacology , Animals , Apoptosis/drug effects , Blood Urea Nitrogen , Creatinine/blood , Creatinine/urine , Male , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Renal Insufficiency/urine , Superoxide Dismutase/metabolism , Water/chemistry
11.
Environ Sci Technol ; 53(12): 6804-6813, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31117546

ABSTRACT

Although the distribution of progestagens in aquatic environments has been widely reported, details on their uptake, elimination, and biotransformation in fish have received little attention. This study investigated the uptake, elimination, and biotransformation potential of a progestagen, cyproterone acetate (CPTA), in Nile tilapia ( Oreochromis niloticus) exposed to an environmentally relevant concentration under semistatic regimes. CPTA in tilapia tissues followed a similar pattern, reaching a concentration plateau within 4 days of exposure, and dropping to below limits of quantitation within 4 days of elimination. The calculated steady-state bioconcentration factors suggest a low bioconcentration potential of CPTA in juvenile tilapia. Results of enzymatic hydrolysis treatments revealed that no conjugates of CPTA were present in tissues, but conjugated biotransformation products of CPTA were found in bile, liver, and muscle. Most CPTA entered tissues and then was biotransformed into seven different products by phase I and phase II metabolism. The concentrations of endogenous cortisol were significantly influenced by CPTA in plasma and liver during the uptake period. These findings suggest that biotransformation products of CPTA should be considered for the assessment of the bioconcentration potential and ecological effects of progestagens.


Subject(s)
Tilapia , Water Pollutants, Chemical , Animals , Biotransformation , Cyproterone Acetate , Progestins
12.
Sci Total Environ ; 666: 894-901, 2019 May 20.
Article in English | MEDLINE | ID: mdl-30818213

ABSTRACT

Great amounts of nutrients discharged into the urbanized coastal areas, which are continuously subject to violently anthropogenic metal contamination, will result in eutrophication and hypoxic episode. In order to study the effects of dissolved oxygen (DO), salinity, nitrogen and phosphorus on the release of six metals including Zn, Pb, Cd, Cu, As and Cr from coastal sediments, a series of 60-days microcosm experiments consisting of sediments and seawater were conducted. Severe hypoxia could result in the enhanced peak values of Pb, Cd, Cu and Cr concentrations in the overlying water. A higher level of water salinity could elevate the peak value of As concentration in water column, and a higher level of nitrogen could increase the peak value of Zn concentration in water. The exchange fluxes demonstrated that the diffusion from the sediments was a dominant process during the first 10 days, However, a relative equilibrium of adsorption and precipitation in the sediment-water interface reached during the later periods. In addition, the bioavailability of the studied metals in sediments was elevated under severe hypoxia, or a high level of water salinity, or high levels of nitrogen and phosphorus. The results of linear regression analysis suggested that higher metal bioavailability in sediments could facilitate the metal release, but the process could be restrained by the higher aqueous phosphorus due to the precipitation of metal phosphates.

13.
Sci Total Environ ; 650(Pt 2): 2221-2230, 2019 Feb 10.
Article in English | MEDLINE | ID: mdl-30292115

ABSTRACT

In-situ study on arsenic speciation and the release kinetics in marine sediments was scarce. In this study, the distributions of labile As and their speciation in coastal sediments of Daya Bay were obtained by separate diffusive gradients in thin films (DGT) probes. Results showed that the DGT-labile As(V) was the main speciation in surface sediments (from -20 to 0 mm) with a concentration range of 0.07-3.05 µg·L-1, while the labile As(III) was the main speciation in deep layers of sediments (from -100 to -20 mm). In coastal areas, mariculture farms was the most dominated contributor to As(V) contamination in surface sediments. Both the apparent diffusion flux estimation and the DGT induced flux in sediments (DIFS) simulation indicated that As(V) contamination in surface sediments of mariculture, harbor and petrochemical areas suffered the potential risk of As(V) release into the overlying water from sediments. DIFS modeling also found that the sediments of mariculture farms were the main sediment As pools. Linear regression analysis indicated that the mobility of As mainly attributed to the As(V) in sediments.

14.
Mar Pollut Bull ; 128: 132-139, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29571356

ABSTRACT

Contamination level, chemical fraction and ecological risk of heavy metals in sediments from Daya Bay (DYB) were conducted in this study. The results revealed that the concentration of Cr, Cu, Zn, As, Cd and Pb in sediments were in the range of 36.38-90.33, 9.54-61.32, 33.54-207.33, 7.80-18.43, 0.13-0.43 and 15.89-30.01 mg kg-1, respectively, with bioavailable fractions of 13.29, 54.16, 47.60, 32.74, 68.14, 26.59%, respectively. A modified potential ecological risk index (MRI) was used for the ecological risk assessment, with ecological risk contribution ratios of 75.73, 14.29, 5.47, 1.74, 1.57 and 1.21% for Cd, As, Cu, Cr, Pb and Zn, respectively. The main contaminants were Cd and As, with their ecological risks "High" and "Moderate" levels, and their enrichment degrees "Moderately Severe" and "Moderate", respectively. The multivariate statistical analysis suggested that the various anthropogenic activities along the bay might contribute mainly to the heavy metals contamination in DYB.


Subject(s)
Bays/chemistry , Environmental Monitoring/methods , Geologic Sediments/chemistry , Metals, Heavy/analysis , Water Pollutants, Chemical/analysis , Chemical Fractionation , China , Human Activities , Multivariate Analysis , Risk Assessment
15.
Biochim Biophys Acta Mol Basis Dis ; 1864(4 Pt A): 1037-1050, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29309922

ABSTRACT

Obesity, a major contributor to the development of cardiovascular diseases, is associated with an autonomic imbalance characterized by sympathetic hyperactivity and diminished vagal activity. Vagal activation plays important roles in weight loss and improvement of cardiac function. Pyridostigmine is a reversible acetylcholinesterase inhibitor, but whether it ameliorates cardiac lipid accumulation and cardiac remodeling in rats fed a high-fat diet has not been determined. This study investigated the effects of pyridostigmine on high-fat diet-induced cardiac dysfunction and explored the potential mechanisms. Rats were fed a normal or high-fat diet and treated with pyridostigmine. Vagal discharge was evaluated using the BL-420S system, and cardiac function by echocardiograms. Lipid deposition and cardiac remodeling were determined histologically. Lipid utility was assessed by qPCR. A high-fat diet led to a significant reduction in vagal discharge and lipid utility and a marked increase in lipid accumulation, cardiac remodeling, and cardiac dysfunction. Pyridostigmine improved vagal activity and lipid metabolism disorder and cardiac remodeling, accompanied by an improvement of cardiac function in high-fat diet-fed rats. An increase in the browning of white adipose tissue in pyridostigmine-treated rats was also observed and linked to the expression of UCP-1 and CIDEA. Additionally, pyridostigmine facilitated activation of brown adipose tissue via activation of the SIRT-1/AMPK/PGC-1α pathway. In conclusion, a high-fat diet resulted in cardiac lipid accumulation, cardiac remodeling, and a significant decrease in vagal discharge. Pyridostigmine ameliorated cardiomyopathy, an effect related to reduced cardiac lipid accumulation, and facilitated the browning of white adipose tissue while activating brown adipose tissue.


Subject(s)
Adipose Tissue/metabolism , Cardiomyopathies/prevention & control , Dietary Fats/adverse effects , Pyridostigmine Bromide/pharmacology , Vagus Nerve/physiopathology , Adipose Tissue/pathology , Adipose Tissue/physiopathology , Animals , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Cardiomyopathies/physiopathology , Dietary Fats/pharmacology , Lipid Metabolism/drug effects , Male , Muscle Proteins/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Vagus Nerve/metabolism , Vagus Nerve/pathology
16.
Mol Med Rep ; 17(2): 3021-3027, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29257242

ABSTRACT

MicroRNA (miR)­138 serves an important role in the proliferation, differentiation and apoptosis of human pulmonary artery smooth muscle cells (HPASMCs), indi-cating the involvement of miR­138 in the development and progression of pulmonary artery hypertension (PAH). Potassium channel subfamily K member 3 (TASK­1), a two­pore domain K+ channel, is expressed in HPASMCs and is associated with hypoxic PAH. However, whether miR­138 mediates PAH through targeting TASK­1 is not known. In the present study, HPASMCs were transfected with miR­138 mimic to establish a PAH model in vitro, and the effects of a miR­138 inhibitor and a TASK­1 inhibitor (A293) were examined. Cell proliferation and mitochondrial membrane potential (MMP) were measured by CCK­8 assay and flow cytometry, respectively. Reverse transcription-quantitative polymerase chain reaction and western blotting were performed to examine the expression of miR­138, TASK­1, Bcl­2, caspase­3 and activation of extracellular signal­regulated kinase 1/2 (ERK1/2). A dual­luciferase reporter assay was also used to analyse the expression level of TASK­1 in HPASMCs. The results of the present study demonstrated that the miR­138 mimic promoted proliferation and MMP level, which was similar to the effect of A293 treatment on HPASMCs. However, the miR­138 inhibitor inhibited the effects induced by miR­138 mimic or A293 treatment, as demonstrated by a decrease in proliferation and MMP level in HPASMCs, accompanied by a decrease of Bcl­2 and an increase of caspase­3 expression levels, as well as ERK1/2 activation. The dual­luciferase reporter assay indicated that TASK­1 expression was negatively regulated by miR­138. The results of the present study suggested that miR­138 promoted proliferation and suppressed mitochondrial depolarization of HPASMCs by targeting TASK­1.


Subject(s)
Cell Proliferation , Membrane Potential, Mitochondrial , MicroRNAs/genetics , Myocytes, Smooth Muscle/cytology , Nerve Tissue Proteins/genetics , Potassium Channels, Tandem Pore Domain/genetics , Pulmonary Artery/cytology , Aged , Cells, Cultured , Female , Gene Expression Regulation , Humans , MAP Kinase Signaling System , Male , Middle Aged , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/metabolism
17.
Toxicol Lett ; 281: 1-9, 2017 Nov 05.
Article in English | MEDLINE | ID: mdl-28844481

ABSTRACT

Excessive oxidative stress and inflammation in hypothalamic paraventricular nucleus (PVN) are implicated in the pathogenesis of hypertension. It is reported that tert-butylhydroquinone (tBHQ), a nuclear factor erythroid 2-related factor 2(Nrf2)-inducer, has a variety of pharmacological activities such as anti-oxidation and anti-inflammatory effect. The objective of this study was to investigate the effects of tBHQ in high salt induced hypertension and to identify whether the beneficial effects were induced by inhibiting PVN oxidative stress and inflammation. Male Sprague-Dawley rats were fed with high salt diet (HS, 8% NaCl) or normal salt diet (NS, 0.3% NaCl). These rats were administration of tBHQ (150mg/kg/d) by oral gavage for 16 weeks. Our results showed that high salt intake resulted in higher mean arterial pressure, cardiac hypertrophy as well as increased plasma level of norepinephrine and interleukin (IL)-1ß, IL-6 compared with NS rats. It increased PVN level of reactive oxygen species, gp91phox, IL-1ß, IL-6, p-IKKß and nuclear factor-kappa B (NF-κB) activity, decreased PVN level of Nrf2 and Cu/Zn-SOD. Chronic administration of tBHQ significantly attenuated these changes in HS rats. These data suggest that the protective effects of tBHQ in salt induced hypertension are partly due to inhibiting oxidative stress and inflammation in PVN.


Subject(s)
Hydroquinones/pharmacology , Hypertension/drug therapy , Inflammation/drug therapy , Oxidative Stress/drug effects , Paraventricular Hypothalamic Nucleus/drug effects , Sodium Chloride, Dietary/adverse effects , Animals , Antioxidants/pharmacology , Arterial Pressure , Disease Models, Animal , Hypertension/chemically induced , Interleukin-1beta/blood , Interleukin-6/blood , Male , Membrane Glycoproteins/blood , NADPH Oxidase 2 , NADPH Oxidases/blood , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Norepinephrine/blood , Paraventricular Hypothalamic Nucleus/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Sodium Chloride, Dietary/administration & dosage , Superoxide Dismutase/metabolism
18.
Clin Exp Pharmacol Physiol ; 44(12): 1192-1200, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28732106

ABSTRACT

It is well-accepted that inflammation plays an important role in the development of cardiac remodelling and that therapeutic approaches targeting inflammation can inhibit cardiac remodelling. Although a large amount of evidence indicates that activation of α7 nicotinic acetylcholine receptor (α7nAChR) causes an anti-inflammatory effect, the role of α7nAChR in cardiac remodelling and the underlying mechanism have not been established. To investigate the effect of the specific α7nAChR agonist, PNU282987, on cardiac remodelling induced by isoproterenol (ISO 60 mg/kg per day) in mice, the cardiomyocyte cross-sectional area (CSA) and collagen volume fraction were evaluated by hematoxylin and eosin (HE) and Masson staining, respectively. Cardiac function and ventricular wall thickness were measured by echocardiography. The protein expressions of collagen I, matrix metalloproteinase 9 (MMP-9), transforming growth factor ß1 (TGF-ß1), and Smad3 were analyzed by Western blot. ISO-induced cardiac hypertrophy, characterized by an increase in the heart weight/body weight ratio, CSA and ventricular wall thickness. Moreover, cardiac fibrosis indices, such as collagen volume fraction, MMP-9 and collagen I protein expression, were also increased by ISO. PNU282987 not only attenuated cardiac hypertrophy but also decreased the cardiac fibrosis induced by ISO. Furthermore, PNU282987 suppressed TGF-ß1 protein expression and the phosphorylation of Smad3 induced by ISO. In conclusion, PNU282987 ameliorated the cardiac remodelling induced by ISO, which may be related to the TGF-ß1/Smad3 pathway. These data imply that the α7nAChR may represent a novel therapeutic target for cardiac remodelling in many cardiovascular diseases.


Subject(s)
Benzamides/therapeutic use , Bridged Bicyclo Compounds/therapeutic use , Cardiomegaly/drug therapy , Nicotinic Agonists/therapeutic use , Smad3 Protein/metabolism , Transforming Growth Factor beta1/metabolism , Ventricular Remodeling/drug effects , alpha7 Nicotinic Acetylcholine Receptor/agonists , Animals , Benzamides/administration & dosage , Bridged Bicyclo Compounds/administration & dosage , Cardiomegaly/metabolism , Cardiomegaly/pathology , Isoproterenol/pharmacology , Male , Mice, Inbred BALB C , Myocardium/metabolism , Myocardium/pathology , Nicotinic Agonists/administration & dosage , Signal Transduction
19.
Sci Rep ; 7: 43107, 2017 03 24.
Article in English | MEDLINE | ID: mdl-28338001

ABSTRACT

Brain renin-angiotensin system (RAS) could regulate oxidative stress in the paraventricular nucleus (PVN) in the development of hypertension. This study was designed to explore the precise mechanisms of RAS acting on reactive oxygen species (ROS) in salt-induced hypertension. Male Wistar rats were administered with a high-salt diet (HS, 8.0% NaCl) for 8 weeks to induced hypertension. Those rats were received PVN infusion of AT1R antagonist losartan (LOS, 10 µg/h) or microinjection of small interfering RNAs for protein kinase C γ (PKCγ siRNA) once a day for 2 weeks. High salt intake resulted in higher levels of AT1R, PKCγ, Rac1 activity, superoxide and malondialdehyde (MDA) activity, but lower levels of copper/zinc superoxide dismutase (Cu/Zn-SOD), superoxide dismutase (SOD) and glutathione (GSH) in PVN than control animals. PVN infusion of LOS not only attenuated the PVN levels of AT1R, PKCγ, Rac1 activity, superoxide and decreased the arterial pressure, but also increased the PVN antioxidant capacity in hypertension. PVN microinjection of PKCγ siRNA had the same effect on LOS above responses to hypertension but no effect on PVN level of AT1R. These results, for the first time, identified that the precise signaling pathway of RAS regulating ROS in PVN is via AT1R/PKCγ/Rac1 in salt-induced hypertension.


Subject(s)
Hypertension/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Reactive Oxygen Species/metabolism , Renin-Angiotensin System , Signal Transduction , Animals , Hypertension/chemically induced , Hypertension/physiopathology , Male , Paraventricular Hypothalamic Nucleus/physiopathology , Protein Kinase C/metabolism , Rats , Rats, Sprague-Dawley , Sodium Chloride, Dietary/adverse effects , rac1 GTP-Binding Protein/metabolism
20.
J Cell Mol Med ; 21(9): 2106-2116, 2017 09.
Article in English | MEDLINE | ID: mdl-28296184

ABSTRACT

Cardiac hypertrophy is associated with autonomic imbalance, characterized by enhanced sympathetic activity and withdrawal of parasympathetic control. Increased parasympathetic function improves ventricular performance. However, whether pyridostigmine, a reversible acetylcholinesterase inhibitor, can offset cardiac hypertrophy induced by pressure overload remains unclear. Hence, this study aimed to determine whether pyridostigmine can ameliorate pressure overload-induced cardiac hypertrophy and identify the underlying mechanisms. Rats were subjected to either sham or constriction of abdominal aorta surgery and treated with or without pyridostigmine for 8 weeks. Vagal activity and cardiac function were determined using PowerLab. Cardiac hypertrophy was evaluated using various histological stains. Protein markers for cardiac hypertrophy were quantitated by Western blot and immunoprecipitation. Pressure overload resulted in a marked reduction in vagal discharge and a profound increase in cardiac hypertrophy index and cardiac dysfunction. Pyridostigmine increased the acetylcholine levels by inhibiting acetylcholinesterase in rats with pressure overload. Pyridostigmine significantly attenuated cardiac hypertrophy based on reduction in left ventricular weight/body weight, suppression of the levels of atrial natriuretic peptide, brain natriuretic peptide and ß-myosin heavy chain, and a reduction in cardiac fibrosis. These effects were accompanied by marked improvement of cardiac function. Additionally, pyridostigmine inhibited the CaN/NFAT3/GATA4 pathway and suppressed Orai1/STIM1 complex formation. In conclusion, pressure overload resulted in cardiac hypertrophy, cardiac dysfunction and a significant reduction in vagal discharge. Pyridostigmine attenuated cardiac hypertrophy and improved cardiac function, which was related to improved cholinergic transmission efficiency (decreased acetylcholinesterase and increased acetylcholine), inhibition of the CaN/NFAT3/GATA4 pathway and suppression of the interaction of Orai1/STIM1.


Subject(s)
Calcineurin/metabolism , Cardiomegaly/drug therapy , Cardiomegaly/metabolism , Pressure , Pyridostigmine Bromide/administration & dosage , Pyridostigmine Bromide/therapeutic use , Signal Transduction , Animals , Cardiomegaly/diagnostic imaging , Cardiomegaly/physiopathology , GATA4 Transcription Factor/metabolism , Heart Function Tests , Hemodynamics/drug effects , Male , NFATC Transcription Factors/metabolism , ORAI1 Protein/metabolism , Protein Binding/drug effects , Rats, Sprague-Dawley , Stromal Interaction Molecule 1/metabolism , Time Factors , Vagus Nerve/drug effects , Vagus Nerve/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...