Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
1.
Int Immunopharmacol ; 137: 112373, 2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38852523

ABSTRACT

Although penehyclidine hydrochloride (PHC) has been identified to alleviate myocardial injury induced by ischemia/reperfusion (I/R), the regulatory molecules and related mechanisms are unknown. In this study, bioinformatics, molecular biology, and biochemistry methods were used to explore the molecular mechanisms and targets of PHC. In the myocardial ischemia-reperfusion injury (MIRI)-induced rat model, PHC pretreatment significantly improved cardiac function (p < 0.01). Multiple differentially expressed genes, including Z-DNA binding protein 1 (ZBP1), were identified through mRNA sequencing analysis of myocardial ischemic penumbra tissue in MIRI rats. The transduction of the ZBP1 adenovirus vector (Ad-Zbp1) in PHC-pretreated rats exhibited a reversible augmentation in myocardial infarct size (p < 0.01), pronounced pathological damage to the myocardial tissue, as well as a significant elevation of serum myocardial enzymes (p < 0.05). The interaction among ZBP1, fas-associating via death domain (FADD), and receptor-interacting serine/threonine-protein kinase 3 (RIPK3) leads to a remarkable up-regulation of cleaved-Caspase-1 (Cl-Casp-1), N-terminal gasdermin D (N-GSDMD), phospho-mixed lineage kinase domain-like Ser358 (p-MLKLS358), and other regulatory proteins, thereby triggering pyroptosis, apoptosis, and necroptosis (PANoptosis) in cardiomyocytes of MIRI rats. Moreover, the transduction of Ad-Zbp1 in the oxygen-glucose deprivation/re-oxygenation (OGD/R)-induced H9c2 cell model also dramatically augmented the number of cell deaths. However, the intervention of PHC considerably enhanced cell viability (p < 0.01), effectively mitigated the release of myocardial enzymes (p < 0.05), and markedly attenuated the expression levels of PANoptosis regulatory proteins through restraint of ZBP1 expression. Therefore, the therapeutic efficacy of PHC in improving MIRI might be attributed to targeting ZBP1-mediated PANoptosis.

2.
BMC Pulm Med ; 24(1): 207, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38671448

ABSTRACT

OBJECTIVE: The aim of this research was to examine how penehyclidine hydrochloride (PHC) impacts the occurrence of pyroptosis in lung tissue cells within a rat model of lung ischemia-reperfusion injury. METHODS: Twenty-four Sprague Dawley (SD) rats, weighing 250 g to 270 g, were randomly distributed into three distinct groups as outlined below: a sham operation group (S group), a control group (C group), and a test group (PHC group). Rats in the PHC group received a preliminary intravenous injection of PHC at a dose of 3 mg/kg. At the conclusion of the experiment, lung tissue and blood samples were collected and properly stored for subsequent analysis. The levels of malondialdehyde, superoxide dismutase, and myeloperoxidase in the lung tissue, as well as IL-18 and IL-1ß in the blood serum, were assessed using an Elisa kit. Pyroptosis-related proteins, including Caspase1 p20, GSDMD-N, and NLRP3, were detected through the western blot method. Additionally, the dry-to-wet ratio (D/W) of the lung tissue and the findings from the blood gas analysis were also documented. RESULTS: In contrast to the control group, the PHC group showed enhancements in oxygenation metrics, reductions in oxidative stress and inflammatory reactions, and a decrease in lung injury. Additionally, the PHC group exhibited lowered levels of pyroptosis-associated proteins, including the N-terminal segment of gasdermin D (GSDMD-N), caspase-1p20, and nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3). CONCLUSION: Pre-administration of PHC has the potential to mitigate lung ischemia-reperfusion injuries by suppressing the pyroptosis of lung tissue cells, diminishing inflammatory reactions, and enhancing lung function. The primary mechanism behind anti-pyroptotic effect of PHC appears to involve the inhibition of oxidative stress.


Subject(s)
Gasdermins , Lung , Pyroptosis , Quinuclidines , Rats, Sprague-Dawley , Reperfusion Injury , Animals , Pyroptosis/drug effects , Reperfusion Injury/drug therapy , Reperfusion Injury/prevention & control , Rats , Quinuclidines/pharmacology , Lung/drug effects , Lung/pathology , Lung/metabolism , Male , Malondialdehyde/metabolism , Disease Models, Animal , Interleukin-1beta/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Interleukin-18/metabolism , Phosphate-Binding Proteins/metabolism , Superoxide Dismutase/metabolism , Peroxidase/metabolism , Oxidative Stress/drug effects , Caspase 1/metabolism , Lung Injury/drug therapy , Lung Injury/metabolism
3.
Adv Clin Exp Med ; 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38530320

ABSTRACT

BACKGROUND: Inflammation is a biological response of the immune system to harmful stimuli. Penehyclidine hydrochloride (PCH) can alleviate inflammation and oxidative stress by activating reactive oxygen species (ROS), nuclear factor erythroid 2-related factor (Nrf2) and heme oxygenase 1 (HO-1) in animal models, but there is a lack of cellular evidence. OBJECTIVES: This study investigated the effects of PHC on lipopolysaccharide (LPS)-induced inflammation response and oxidative stress in RAW264.7 cells. MATERIAL AND METHODS: RAW264.7 cells were treated with 1 µg/mL or 5 µg/mL of PHC, with interleukin 6 (IL-6), tumor necrosis factor alpha (TNF-α), IL-1ß, and prostaglandin E2 (PGE2) levels measured with enzyme-linked immunosorbent assay (ELISA) and nitric oxide (NO) measured using the Griess test. Reactive oxygen species were examined with flow cytometry and immunofluorescence, and b-related factor 2 (BRF-2) and NAD(P)H-quinone oxidoreductase 1 (NQO1) using western blot. RESULTS: Penehyclidine hydrochloride partly, but substantially, reversed LPS-related NO and PGE2 production by RAW264.7 cells in a dose-dependent manner and suppressed LPS-induced expression of IL-6, TNF-α and IL-1ß messenger ribonucleic acid (mRNA), secretion of IL-6, TNF-α and IL-1ß, and ROS production. Lipopolysaccharide stimulation did not affect Nrf2, heme oxygenase 1 (HO-1) or NQO1 protein expression in RWA264.7 cells not treated with PHC. However, PHC treatment significantly elevated Nrf2, HO-1 and NQO1 protein in LPS-treated RWA264.7 cells, an effect that was dose-dependent. The ROS scavenging using N-acetyl-L-cysteine abolished the PHC-induced upregulation of Nrf2 and HO-1. CONCLUSIONS: Penehyclidine hydrochloride may alleviate LPS-induced inflammation and oxidative stress by activating Nrf2 signaling in RAW264.7 macrophages. These findings suggest that PHC could alleviate inflammation by targeting activated macrophages.

4.
J Cardiovasc Transl Res ; 16(6): 1373-1382, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37584895

ABSTRACT

Penehyclidine hydrochloride (PHC) is an anticholinergic drug with cardioprotective effects. Ferroptosis is closely related to myocardial ischaemia-reperfusion injury (MIRI). In the present study, MIRI was induced in rats by left anterior descending coronary artery ligation. PHC pretreatment increased haemodynamic parameters and histopathological damage and reduced myocardial infarction size in the MIRI model. PHC pretreatment also inhibited ferroptosis, which was characterized by the decreased levels of Fe2+, 4-hydroxynonenal and ACSL4, and increased levels of GPX4, GSH-Px and GST. In response to 6 h of oxygen-glucose deprivation and 18 h of reoxygenation, PHC pretreatment had the same effects on these factors in H9c2 cells and reduced lipid ROS levels. Furthermore, ACSL4 overexpression reversed the protective effects of PHC on H9c2 cells. These results indicated that PHC inhibited MIRI through ACSL4-mediated ferroptosis. This study demonstrated that PHC could inhibit ferroptosis in MIRI and the relationship among PHC, ACSL4, ferroptosis and MIRI. This study demonstrated the inhibitory effect of PHC on ferroptosis and showed that PHC affects MIRI through ACSL4-mediated ferroptosis in vivo and in vitro.


Subject(s)
Ferroptosis , Myocardial Reperfusion Injury , Rats , Animals , Myocardial Reperfusion Injury/pathology , Cardiotonic Agents , Quinuclidines/pharmacology , Quinuclidines/therapeutic use
5.
BMC Anesthesiol ; 23(1): 237, 2023 07 13.
Article in English | MEDLINE | ID: mdl-37442959

ABSTRACT

AIM: To investigate the effects of penehyclidine hydrochloride combined with dexmedetomidine on pulmonary function in patients undergoing heart valve surgery with cardiopulmonary bypass (CPB). METHODS: A total of 180 patients undergoing elective heart valve surgery with CPB were randomly divided into four groups: 45 in group P (intravenous penehyclidine hydrochloride 0.02 mg/kg 10 min before anesthesia induction and at the beginning of CPB, total 0.04 mg/kg); 43 in group D (dexmedetomidine 0.5 µg/kg/h after induction of anesthesia until the end of anesthesia); 44 in group PD ( penehyclidine hydrochloride 0.04 mg/kg combined with dexmedetomidine 0.5 µg/kg/h intravenously during anesthesia); and 43 in group C (same amount of normal saline 10 min before and after anesthesia induction, to the end of anesthesia, and at the beginning of CPB). The main outcomes were the incidence and severity of postoperative pulmonary complications (PPCs). The secondary outcomes were: (1) extubation time, length of stay in intensive care, and postoperative hospital stay, and adverse events; and (2) pulmonary function evaluation indices (oxygenation index and respiratory index) and plasma inflammatory factor concentrations (tumor necrosis factor-α, interleukin-6, C-reactive protein and procalcitonin) during the perioperative period. RESULTS: The incidence of PPCs in groups P, D and PD after CPB was lower than that in group C (P < 0.05), and the incidence in group PD was significantly lower than that in groups P and D (P < 0.05). The scores for PPCs in groups P, D and PD were lower than those in group C (P < 0.05). CONCLUSION: Combined use of penehyclidine hydrochloride and dexmedetomidine during anesthesia reduced the occurrence of postoperative pulmonary dysfunction, and improved the prognosis of patients undergoing heart valve surgery with CPB. TRIAL REGISTRATION: The trial was registered in the Chinese Clinical Trial Registry on 3/11/2020 (Registration No.: ChiCTR2000039610).


Subject(s)
Dexmedetomidine , Humans , Quinuclidines/therapeutic use , Double-Blind Method , Heart Valves
6.
Int J Immunopathol Pharmacol ; 37: 3946320231192175, 2023.
Article in English | MEDLINE | ID: mdl-37500500

ABSTRACT

INTRODUCTION: Acute lung injury (ALI) attracted attention among physicians because of its high mortality. We aimed to determine whether the phosphatidylinositol-3 kinase (PI3K)/protein kinase B (Akt) pathway is involved in the protective effects of penehyclidine hydrochloride (PHC) against lipopolysaccharide (LPS)-induced ALI. METHODS: H&E staining was used to observed pathological changes in the lung tissues. ELISA was used to evaluate the concentration of inflammatory mediators in the bronchoalveolar lavage fluid (BALF). White-light microscopy was performed to observe the TUNEL-positive nuclei. The viability of NR8383 alveolar macrophages was determined by using CCK-8. The levels of MPO, MDA, SOD, and GSH-Px were analyzed using ELISA kits. Western blotting was used to evaluate the ERS-associated protein levels and the phosphorylation of PI3K and Akt. RESULTS: PHC administration defended against LPS-induced histopathological deterioration and increased pulmonary edema and lung injury scores, while all of these beneficial effects were inhibited by LY. In addition, PHC administration mitigated oxidative stress as indicated by decreases in lung myeloperoxidase (MPO) and malondialdehyde (MDA) concentrations, and increases in glutathione peroxidase (GSH-Px) and superoxide dismutase (SOD) concentrations. It also alleviated LPS-induced inflammation. PHC administration attenuated apoptosis-associated protein levels, improved cell viability, and decreased the number of TdT-mediated dUTP Nick-End Labeling (TUNEL)-positive cells. Furthermore, PHC inhibited ERS-associated protein levels. Meanwhile, the protection of PHC against inflammation, oxidative stress, apoptosis, and ERS was inhibited by LY. Moreover, PHC administration increased PI3K and Akt phosphorylation, indicating that the upregulation of the PI3K/Akt pathway, while this pathway was inhibited by LY. CONCLUSION: PHC significantly activates the PI3K/Akt pathway to ameliorate the extent of damage to pulmonary tissue, inflammation, oxidative stress, apoptosis, and ERS in LPS-induced ALI.


Subject(s)
Acute Lung Injury , Lipopolysaccharides , Rats , Animals , Lipopolysaccharides/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Phosphatidylinositol 3-Kinase/pharmacology , Rats, Sprague-Dawley , Acute Lung Injury/chemically induced , Acute Lung Injury/drug therapy , Acute Lung Injury/prevention & control , Lung , Inflammation/metabolism , Superoxide Dismutase
7.
J Intensive Med ; 3(2): 171-184, 2023 Apr 30.
Article in English | MEDLINE | ID: mdl-37188113

ABSTRACT

Background: Penehyclidine hydrochloride (PHC) has been used for many years as an anticholinergic drug for the treatment of acute organophosphorus pesticide poisoning (AOPP). The purpose of this meta-analysis was to explore whether PHC has advantages over atropine in the use of anticholinergic drugs in AOPP. Methods: We searched Scopus, Embase, Cochrane, PubMed, ProQuest, Ovid, Web of Science, China Science and Technology Journal Database (VIP), Duxiu, Chinese Biomedical literature (CBM), WanFang, and Chinese National Knowledge Infrastructure (CNKI), from inception to March 2022. After all qualified randomized controlled trials (RCTs) were included, we conducted quality evaluation, data extraction, and statistical analysis. Statistics using risk ratios (RR), weighted mean difference (WMD), and standard mean difference (SMD). Results: Our meta-analysis included 20,797 subjects from 240 studies across 242 different hospitals in China. Compared with the atropine group, the PHC group showed decreased mortality rate (RR=0.20, 95% confidence intervals [CI]: 0.16-0.25, P <0.001), hospitalization time (WMD=-3.89, 95% CI: -4.37 to -3.41, P <0.001), overall incidence rate of complications (RR=0.35, 95% CI: 0.28-0.43, P <0.001), overall incidence of adverse reactions (RR=0.19, 95% CI: 0.17-0.22, P <0.001), total symptom disappearance time (SMD=-2.13, 95% CI: -2.35 to -1.90, P <0.001), time for cholinesterase activity to return to normal value 50-60% (SMD=-1.87, 95% CI: -2.03 to -1.70, P <0.001), coma time (WMD=-5.57, 95% CI: -7.20 to -3.95, P <0.001), and mechanical ventilation time (WMD=-2.16, 95% CI: -2.79 to -1.53, P <0.001). Conclusion: PHC has several advantages over atropine as an anticholinergic drug in AOPP.

8.
BMC Anesthesiol ; 23(1): 135, 2023 04 24.
Article in English | MEDLINE | ID: mdl-37095439

ABSTRACT

BACKGROUND: Postoperative nausea and vomiting (PONV) is a common and distressing complication of laparoscopic bariatric surgery (LBS). Penehyclidine hydrochloride has been reported to be effective in preventing PONV. Considering the potential preventive effects of penehyclidine against PONV, we hypothesized that intravenous infusion of penehyclidine may alleviate PONV within the first 48 h in patients scheduled for LBS. METHODS: Patients who underwent LBS were randomly assigned (1:2) to receive saline (Control group, n = 113) or a single intravenous dose of penehyclidine 0.5 mg (PHC group, n = 221). The primary outcome was incidence of PONV within the first 48 h postoperatively. Secondary endpoints included severity of PONV, need for rescue antiemetic therapy, volume of water intake, and time to first flatus. RESULTS: PONV occurred in 159 (48%) patients within the first 48 h postoperatively, including 51% in the Control group and 46% in the PHC group. There was no significant difference in the incidence or severity of PONV between the two groups (P > 0.05). Within the first 24 h and 24-48 h, no significant difference was found in incidence or severity of PONV, postoperative nausea, postoperative vomiting, need for rescue antiemetic therapy, or volume of water intake (P > 0.05). Kaplan-Meier curves showed that penehyclidine was significantly associated with a prolonged time to first flatus (median onset time: 22 h vs. 21 h, P = 0.036). CONCLUSIONS: Penehyclidine did not decrease incidence and severity of PONV in patients undergoing LBS. However, a single intravenous dose of penehyclidine (0.5 mg) was associated with a slightly prolonged time to first flatus. TRIAL REGISTRATION: Chinese Clinical Trial Registry (ChiCTR2100052418, http://www.chictr.org.cn/showprojen.aspx?proj=134893 , date of registration: 25/10/2021).


Subject(s)
Antiemetics , Bariatric Surgery , Laparoscopy , Humans , Postoperative Nausea and Vomiting/drug therapy , Antiemetics/therapeutic use , Flatulence/drug therapy , Double-Blind Method
9.
Comb Chem High Throughput Screen ; 26(6): 1204-1213, 2023.
Article in English | MEDLINE | ID: mdl-35838232

ABSTRACT

AIMS AND OBJECTIVE: The lack of effective treatments for myocardial ischemiareperfusion (MI-R) injury severely restricts the effectiveness of the treatment of ischemic heart disease. In the present research, we aimed to investigate the protective effect and molecular mechanism of penehyclidine hydrochloride (PHC) on MI-R cells. METHODS: Cell viability was quantified using CCK8. Cell apoptosis was analyzed using flow cytometry. Western blot and Elisa assays were used for the detection of target proteins. RESULTS: PHC pretreatment attenuated the inhibition of cell viability and decreased the percentage of apoptosis induced by simulated ischemia reperfusion (SIR). Platelet-derived growth factor B (PDGF-B) and its downstream AKT pathway were activated in PHC pretreated cells. After siRNAPDGF- B transfection, cell viability was inhibited and apoptosis was activated in PHC pretreated SIR cells, suggesting that PHC protected cells from SIR. PDGF-B knockdown also increased the levels of CK, LDH, IL-6 and TNF-α in PHC pretreated SIR cells. The effect of AKT inhibitor on H9C2 cells was consistent with that of PDGF-B knockdown. CONCLUSION: PHC pretreatment can protect cardiomyocytes from the decrease of cell activity and the increase of apoptosis caused by reperfusion through up-regulating PDGF-B to activate PI3K pathway. Our study indicates that PHC is a potential drug to protect cells from reperfusion injury and PDGF-B is a potential target for preventing MI-R injury.


Subject(s)
Myocytes, Cardiac , Reperfusion Injury , Rats , Animals , Myocytes, Cardiac/metabolism , Rats, Wistar , Proto-Oncogene Proteins c-sis/metabolism , Proto-Oncogene Proteins c-sis/pharmacology , Proto-Oncogene Proteins c-sis/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Reperfusion Injury/drug therapy , Ischemia/metabolism , Apoptosis
10.
Zhonghua Lao Dong Wei Sheng Zhi Ye Bing Za Zhi ; 40(10): 779-782, 2022 Oct 20.
Article in Chinese | MEDLINE | ID: mdl-36348562

ABSTRACT

Thiamethoxam belongs to the second generation of neonicotinoid insecticides, and case of acute poisoning with thiamethoxam had never reported in China. This paper reviewed a case of oral poisoning with thiamethoxam pesticides, the patient suffered vomiting, generalized convulsions, confusion, and decreased oxygen saturation. After treated with gastric lavage, ventilator support, and the use of propofol, midazolam, sodium phenobarbital, and sodium valproate, the convulsions could not be controlled. Untill treated with penehyclidine hydrochloride and hemoperfusion combined with hemofiltration, the patient finally recovered and was discharged from the hospital. We suggest that the main treatments for acute severe thiamethoxam poisoning are decontamination and symptomatic support, pentoxifylline hydrochloride and hemoperfusion combined with hemofiltration may improve the patients' prognosis.


Subject(s)
Hemofiltration , Hemoperfusion , Insecticides , Pesticides , Poisoning , Humans , Thiamethoxam , Prognosis , Neonicotinoids , Poisoning/therapy
11.
Bioimpacts ; 12(3): 211-218, 2022.
Article in English | MEDLINE | ID: mdl-35677666

ABSTRACT

Introduction: Penehyclidine hydrochloride (PHC) is an anticholinergic with anti-inflammatory and anti-oxidation activities. PHC displayed protectivity against renal ischemia reperfusion (RIR) injury. Nevertheless, the precise protectivity of PHC on RIR-induced lung injury remains unknown. Methods: We examined the effects of PHC on RIR-induced lung injury and investigated the underlying mechanism. We induced RIR in mice and administrated PHC to RIR mice. Kidney function was monitored by measuring the blood urea nitrogen (BUN) and creatinine level in serum. We evaluated the lung injury, myeloperoxidase (MPO) activity in lung, pro-inflammatory cytokine level, and oxidative markers in serum and lung tissues. We tested the expression level of nuclear factor erythroid 2-related factor 2 (Nrf-2) and heme oxygenase 1 (HO-1) in lung of RIR mice after PHC treatment. Finally, we evaluated the effects of PHC in RIR Nrf2-/- mice. Results: PHC greatly downregulated the serum levels of BUN, creatinine, IL-6, NO, malondialdehyde (MDA), and matrix metalloproteinase-2. PHC also ameliorated the lung injury, decreased the MPO activity, and suppressed production of IL-6, TNF-α, IFN-γ, MDA, and O2-, while it promoted production of superoxide dismutase (SOD) and catalase (CAT) in lung. PHC improved the production of Nrf2 and HO-1. Conclusion: The protectivity of PHC was absent in Nrf2-/- mice. PHC ameliorated RIR-induced lung injury through Nrf2 pathway.

12.
Int Immunopharmacol ; 107: 108680, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35303505

ABSTRACT

BACKGROUND: Diabetic nephropathy (DN) is one of the main complications of diabetes. Penehyclidine hydrochloride (PHC) has anti-inflammatory, anti-apoptotic and anti-oxidative stress effects. Nevertheless, whether PHC can be used to prevent podocyte injury has not been reported. OBJECTIVES: This present study aimed to identify the functional role of PHC in DN as well as its underlying mechanism. METHODS: The high-glucose (HG)-induced podocyte damage in vitro model was established. The proliferation, apoptotic rate, inflammatory factors, and gene/protein expressions of HG-induced MPC5 cells were determined using CCK-8 assay, flow cytometry, ELISA, real-time PCR, and Western blot upon PHC treatment. Co-immunoprecipitation experiments and pull-down assay were performed to verify the interactions between fibrinogen-like protein 2 (Fgl2) and toll-like receptor 4 (TLR4) as well as TLR4 and NLRP3. A rat in vivo model was used to confirm the effect of PHC treatment. RESULTS: PHC treatment reduced Fgl2 expression and inhibited HG-induced podocyte injury and DN-induced kidney damage. Flg2 was associated with TLR4 and NLRP3. It was further proved that PHC treatment suppressed the TLR4-NF-кB and NLRP3-Caspase-1 pathways through Fgl2, which eventually inhibited inflammatory cytokines and prevented HG-induced podocyte injury both in vitro and in vivo. CONCLUSION: PHC treatment possibly ameliorates DN by preventing podocyte injury via inactivating the TLR4-NF-кB and NLRP3-Caspase-1 signaling pathways by Flg2.


Subject(s)
Diabetes Mellitus , Diabetic Nephropathies , Podocytes , Animals , Caspase 1/metabolism , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/metabolism , Female , Fibrinogen/metabolism , Humans , Inflammation/drug therapy , Inflammation/metabolism , Male , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Quinuclidines , Rats , Toll-Like Receptor 4/metabolism
13.
J Invest Surg ; 35(5): 1050-1061, 2022 May.
Article in English | MEDLINE | ID: mdl-34696682

ABSTRACT

Background: The activation of alveolar macrophages (AMs) modulated via leucine-rich repeat (NLR) pyrin domain containing 3 (NLRP3) inflammasome activation is key to the progression of renal ischemia/reperfusion (rI/R)-mediated acute lung injury (ALI). Sirtuin-1 (SIRT1) can attenuate NLRP3 inflammasome activation during I/R stress and may be an important mechanism underlying ALI pathogenesis. Penehyclidine hydrochloride (PHC), an anticholinergic drug, exerts protective effects against rI/R-mediated ALI. This study aimed to decipher the effects of PHC on SIRT1 activation and the underlying mechanism of the protective activity of PHC against rI/R-mediated ALI.Materials and methods: We used an ALI rat model and the rat AMs cell line NR8383 to assess the degree of lung injury in vivo and in vitro.Results: The results show that PHC attenuates rI/R-mediated lung injury indices, myeloperoxidase, and apoptosis in vivo. It decreases the rI/R-mediated release of prostaglandin E2 and nitric oxide, mitochondrial reactive oxygen species production, and the activity of NADPH oxidase-4 in vitro. PHC ameliorates the rI/R-induced activation of the thioredoxin-interacting protein, caspase 1 (P10 unit), and NLRP3 inflammasome, along with reduced activation of interleukin-1ß and interleukin-18 in vitro. We show that PHC alleviates the rI/R-induced reduction of SIRT1 and the depletion of SIRT1 eliminates the ameliorating activity of PHC on the NLRP3 inflammasome activation in vitro. Conclusions: In summary, the findings suggest that PHC ameliorates the rI/R-mediated ALI through the SIRT1-mediated NLRP3 inflammasome activation.


Subject(s)
Acute Lung Injury , Inflammasomes , Acute Lung Injury/metabolism , Animals , Inflammasomes/adverse effects , Inflammasomes/metabolism , Ischemia , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Quinuclidines , Rats , Reperfusion , Sirtuin 1/metabolism
14.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-960447

ABSTRACT

Background The current treatment program with penehyclidine hydrochloride (PHC) for acute severe organophosphorus pesticide poisoning (ASOPP) patients exerts a positive effect but with concerned adverse reactions. Objective To evaluate the treatment effect of a revised ASOPP treatment program with PHC. Methods A prospective single-blind randomized controlled trial was conducted. A total of 157 patients with ASOPP were divided into a revised treatment group (82 cases) and a conventional treatment group (75 cases) by random number table. The two groups received the same basic treatment measures including active life support, routine gastric lavage, catharsis, and pralidoxime treatment. The revised treatment group followed a revised PHC treatment protocol initiated by first a small dose of PHC and followed by small doses of PHC administration/discontinuation through frequent observations at different time points. The conventional treatment group received the conventional program. Treatment effects and incidence rates of possible adverse reactions were compared between the two groups. Results Compared with the conventional treatment group, the revised treatment group obtained delay in penetrogenation time point, higher success rate in catharsis, earlier cholinesterase-turning time, and shorter hospitalization period with statistical significance (all Ps<0.05). No differences were found in terms of time for symptoms of poisoning to disappear, incidence rates of intermediate myasthenic syndrome and delayed polyneuropathy, mechanical ventilation time, and cure rate (all Ps>0.05). Less adverse reactions occurred in the revised treatment group including tachycardia and delirium than in the conventional treatment control group (all Ps<0.05). Conclusion The revised ASOPP treatment program with PHC is similar to the current recommended treatment program in treatment effects, but with less adverse reactions.

15.
Mol Med Rep ; 24(5)2021 11.
Article in English | MEDLINE | ID: mdl-34468006

ABSTRACT

The aim of the present study was to investigate the effect of penehyclidine hydrochloride (PHC) pretreatment on mice with lipopolysaccharide (LPS)­induced acute lung injury (ALI) and its possible underlying mechanisms. Mice were randomly separated into six groups: i) Sham group; ii) LPS group; iii) LPS + PHC group; iv) tumor necrosis factor a­induced protein 8­like protein 2 (TIPE2) group; v) LPS + TIPE2 group; and vi) LPS + TIPE2 + PHC group. The ALI model was induced using LPS through intratracheal injection. The mice received adenovirus gene to induce the overexpression of TIPE2. After mice were sacrificed, lung injury indices were assessed, and arterial blood, bronchoalveolar lavage fluid and lung tissues were collected for subsequent assays. Expression levels of related proteins were detected by using western blotting. It was found that compared with the sham group, the mice treated with LPS showed increased lung injury and dysfunctions of gas exchange. However, these trends were significantly ameliorated in the LPS + PHC group. Evaluation of protein expression in lung tissues showed that the increased expression of nuclear NF­κB p65 and p­c­Jun N­terminal kinase (JNK) induced by LPS were suppressed in the LPS + PHC group and the expression of TIPE2 was increased. The mice that received adenovirus gene to induce TIPE2 overexpression could also showed protective effects compared with the mice in the LPS group. However, the expression of TIPE2 decreased rather than increased in LPS group. In the mice pretreated with PHC, the expression of TIPE2 increased in mice with LPS­induced ALI. To conclude, PHC pretreatment could inhibit the occurrence of inflammation and apoptosis in LPS­induced ALI. This process may be related to the activation of TIPE2 and the inhibition of NF­κB and JNK signaling pathway in the lungs of mice.


Subject(s)
Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lipopolysaccharides/adverse effects , Quinuclidines/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Acute Lung Injury/chemically induced , Acute Lung Injury/pathology , Animals , Apoptosis/drug effects , Bronchoalveolar Lavage Fluid , Disease Models, Animal , Inflammation , Intracellular Signaling Peptides and Proteins/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Lung/pathology , MAP Kinase Signaling System , Male , Mice , Mice, Inbred BALB C , NF-kappa B/metabolism , Transcription Factor RelA/metabolism
16.
Article in English | MEDLINE | ID: mdl-33833508

ABSTRACT

BACKGROUND: Penehyclidine hydrochloride is a selective antagonist of M1 and M3 receptors. Clinical studies suggest that it is a potential drug for the treatment of chronic obstructive pulmonary disease (COPD). The purpose of this study was to evaluate the effect of penehyclidine hydrochloride on the inflammatory response of lung tissue during mechanical ventilation in rats with COPD and explore the role of the c-Jun N-terminal kinase/stress-activated protein kinase (JNK/SAPK) signaling pathway. METHODS: Eight-week-old male Sprague Dawley rats were exposed to cigarette smoke for 30 minutes every day for two months, and on the first and thirtieth days, 200 ug of lipopolysaccharide was injected into the trachea. Two months later, the rats were randomly divided into the control group (C), model group (M), model + normal saline group (N), and penehyclidine hydrochloride group (H) to undergo anesthesia and mechanical ventilation. In group H, 1 mg/kg of penehyclidine hydrochloride was injected intravenously. RESULTS: The results showed that: ① Compared with group C, the other groups all showed typical chronic obstructive pathological changes in the lung tissue; their wet/dry weight ratio (W/D), TNF-α, JNK, and p-JNK levels increased (P < 0.05), and their interleukin (IL)-10 levels decreased (P < 0.05). ② Compared with group M, there was no significant change in the lung tissue indexes in group N (P > 0.05). ③ Compared with group N, the W/D, TNF-α, JNK, and p-JNK levels in group H decreased (P < 0.05), while the levels of IL-10 increased (P < 0.05). CONCLUSION: Penehyclidine hydrochloride can alleviate the pulmonary inflammatory response in rats with COPD undergoing mechanical ventilation. The JNK/SAPK signaling pathway may be involved in this process.


Subject(s)
Pulmonary Disease, Chronic Obstructive , Animals , Lung , Male , Pulmonary Disease, Chronic Obstructive/drug therapy , Quinuclidines , Rats , Rats, Sprague-Dawley , Respiration, Artificial
17.
Mol Med Rep ; 23(2)2021 02.
Article in English | MEDLINE | ID: mdl-33300058

ABSTRACT

Acute progressive hypoxic respiratory failure caused by various predisposing factors is known as acute respiratory distress syndrome (ARDS). Although penehyclidine hydrochloride (PHC), an anticholinergic drug, is widely applied in clinical practice, the specific mechanisms underlying PHC in the treatment of ARDS are not completely understood. In the present study, BEAS­2B cells were treated with 10 ng/ml lipopolysaccharide (LPS) to establish an ARDS cell model and a rat model of acute lung injury (ALI). The influences of PHC and/or autophagy inhibitor (3­methyladenine (3­MA)) on the morphology, autophagy, proliferation and apoptosis of cells and tissues were evaluated using hematoxylin and eosin staining, Cell Counting Kit­8 assays, Hoechst staining, TUNEL staining, flow cytometry, immunofluorescence assays, ELISAs and scanning electron microscopy. The expression levels of apoptosis­ and autophagy­related proteins were measured via western blotting. The results indicated that PHC enhanced proliferation and autophagy, and decreased apoptosis and the inflammatory response in LPS­induced BEAS­2B cells and ALI model rats. In addition, 3­MA reversed the effects of PHC on proliferation, inflammation, apoptosis and autophagy in LPS­induced BEAS­2B cells. Therefore, the present study suggested that PHC demonstrated a protective effect in LPS­induced ARDS by regulating an autophagy­related pathway.


Subject(s)
Autophagy/drug effects , Lipopolysaccharides/toxicity , Quinuclidines/pharmacology , Respiratory Distress Syndrome , Signal Transduction/drug effects , Animals , Cell Line , Humans , Male , Rats , Rats, Sprague-Dawley , Respiratory Distress Syndrome/chemically induced , Respiratory Distress Syndrome/drug therapy , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/pathology
18.
Aging (Albany NY) ; 12(13): 13400-13421, 2020 07 11.
Article in English | MEDLINE | ID: mdl-32652517

ABSTRACT

The nuclear factor (NF)-κB and NOD-like receptor protein 3 (NLRP3) pathways promote inflammatory signaling that injures the kidneys, whereas the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway promotes anti-inflammatory signaling that inhibits oxidative damage. Penehyclidine hydrochloride (PHC) inhibits NF-κB and activates Nrf2 signaling. We investigated whether PHC induces communication between the Nrf2 and NF-κB/NLRP3 pathways, thereby protecting against renal ischemia/reperfusion (rI/R)-induced lung inflammation. Rat alveolar macrophages (NR8383 cells) were stimulated for 24 h with PHC with or without brusatol (a Nrf2 antagonist), after which they were treated for 4 h with tert-butyl hydroperoxide (10 mM). PHC Nrf2-dependently alleviated tert-butyl hydroperoxide-induced reactive oxygen species production in alveolar macrophages. Additionally, wild-type and Nrf2-/- rats were each divided into four groups: (1) sham, (2) PHC (1 mg/kg), (3) rI/R and (4) rI/R + PHC (1 mg/kg). PHC markedly induced the Nrf2 and adenosine monophosphate-activated protein kinase pathways and suppressed rI/R-induced NF-κB and NLRP3 activation in the lungs. Nrf2 deficiency diminished the ability of PHC to ameliorate rI/R-induced histopathological alterations and reactive oxygen species release in the lungs; however, PHC inhibited NLRP3 signaling Nrf2-dependently, while it inhibited NF-κB signaling Nrf2-independently. Our findings demonstrate the beneficial effects of PHC on rI/R-induced lung inflammation.


Subject(s)
Acute Lung Injury/prevention & control , Kidney Diseases/complications , NF-E2-Related Factor 2/metabolism , Quinuclidines/pharmacology , Reperfusion Injury/complications , Acute Lung Injury/etiology , Acute Lung Injury/pathology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Disease Models, Animal , Gene Knockout Techniques , Humans , Kidney/blood supply , Kidney/drug effects , Kidney/pathology , Lung/cytology , Lung/drug effects , Lung/pathology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/pathology , Male , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/genetics , Oxidative Stress/drug effects , Oxidative Stress/genetics , Quassins/pharmacology , Quinuclidines/therapeutic use , Rats , Rats, Transgenic , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , tert-Butylhydroperoxide/toxicity
19.
Cell Biol Int ; 44(6): 1353-1362, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32125033

ABSTRACT

Penehyclidine hydrochloride (PHC) can protect against myocardial ischemia/reperfusion (I/R) injury. However, the possible mechanisms of PHC in anoxia/reoxygenation (A/R)-induced injury in H9c2 cells remain unclear. In the present study, H9c2 cells were pretreated with PI3K/Akt inhibitor LY294002, ATP-sensitive K+ (KATP) channel blocker 5-hydroxydecanoate (5-HD), PHC, or KATP channel opener diazoxide (DZ) before subjecting to A/R injury. Cell viability and cell apoptosis were determined by cell counting kit-8 assay and annexin V/PI assay, respectively. Myocardial injury was evaluated by measuring creatine kinase (CK) and lactate dehydrogenase (LDH) activities. Intracellular Ca2+ levels, reactive oxygen species (ROS) generation, mitochondrial membrane potential (ΔΨm ), and mitochondrial permeability transition pore (mPTP) were measured. The levels of cytoplasmic/mitochondrial cytochrome c (Cyt-C), Bax, Bcl-2, cleaved caspase-3, KATP channel subunits (Kir6.2 and SUR2A), and the members of the Akt/GSK-3ß and Akt/mTOR signaling pathways were determined by western blotting. We found that PHC preconditioning alleviated A/R-induced cell injury by increasing cell viability, reducing CK and LDH activities, and inhibiting cell apoptosis. In addition, PHC preconditioning ameliorated intracellular Ca2+ overload and ROS production, accompanied by inhibition of both mPTP opening and Cyt-C release into cytoplasm, and maintenance of ΔΨm . Moreover, PHC preconditioning activated mitochondrial KATP channels, and modulated the Akt/GSK-3ß and Akt/mTOR signaling pathways. Similar effects were observed upon treatment with DZ. Pretreatment with LY294002 or 5-HD blocked the beneficial effects of PHC. These results suggest that the protective effects of PHC preconditioning on A/R injury may be related to mitochondrial KATP channels, as well as the Akt/GSK-3ß and Akt/mTOR signaling pathways.


Subject(s)
Mitochondria/drug effects , Myocytes, Cardiac/drug effects , Protective Agents/pharmacology , Quinuclidines/pharmacology , Animals , Apoptosis/drug effects , Cell Line , Membrane Potential, Mitochondrial/drug effects , Myocardial Reperfusion Injury/drug therapy , Myocytes, Cardiac/pathology , Potassium Channels/metabolism , Rats , Signal Transduction/drug effects
20.
Mol Med Rep ; 21(5): 2182-2192, 2020 05.
Article in English | MEDLINE | ID: mdl-32186764

ABSTRACT

Penehyclidine hydrochloride (PHC) suppresses renal ischemia and reperfusion (I/R) injury (IRI); however, the underlying mechanism of action that achieves this function remains largely unknown. The present study aimed to investigate the potential role of autophagy in PHC­induced suppression of renal IRI, as well as the involvement of cell proliferation and apoptosis. A rat IRI model and a cellular hypoxia/oxygenation (H/R) model were established; PHC, 3­methyladenine (3­MA) and rapamycin (Rapa) were administered to the IRI model rats prior to I/R induction and to H/R cells following reperfusion. Serum creatinine was measured using a biochemistry analyzer, whereas aspartate aminotransferase (ASAT) and alanine aminotransferase (ALAT) expression levels were detected using ELISA kits. Renal tissue injury was evaluated by histological examination. In addition, microtubule­associated protein light chain 3B (LC3B) expression, autophagosome formation, cell proliferation and apoptosis were detected in the cellular H/R model. The results demonstrated that I/R induced renal injury in IRI model rats, upregulated serum creatinine, ALAT and ASAT expression levels, and increased autophagic processes. In contrast, pretreatment with PHC or Rapa significantly prevented these I/R­induced changes, whereas the administration of 3­MA enhanced I/R­induced injuries through suppressing autophagy. PHC and Rapa increased LC3B and Beclin­1 expression levels, but decreased sequestome 1 (p62) expression in the cellular H/R model, whereas 3­MA prevented these PHC­induced changes. PHC and Rapa promoted proliferation and autophagy in the cellular H/R model; these effects were accompanied by increased expression levels of LC3B and Beclin­1, and reduced p62 expression levels, whereas these levels were inhibited by 3­MA. Furthermore, PHC and Rapa inhibited apoptosis in the cellular H/R model through increasing Bcl­2 expression levels, and suppressing Bax and caspase­3 expression levels; the opposite effect was induced by 3­MA. In conclusion, PHC suppressed renal IRI through the induction of autophagy, which in turn promoted proliferation and suppressed apoptosis in renal cells.


Subject(s)
Autophagy/drug effects , Ischemia/metabolism , Kidney/metabolism , Quinuclidines/adverse effects , Reperfusion Injury/metabolism , Adenine/adverse effects , Adenine/analogs & derivatives , Alanine Transaminase/metabolism , Animals , Apoptosis/drug effects , Aspartate Aminotransferases/metabolism , Beclin-1/metabolism , Caspase 3/metabolism , Cell Proliferation/drug effects , Disease Models, Animal , Ischemia/chemically induced , Kidney/injuries , Kidney/pathology , Male , Rats , Rats, Sprague-Dawley , Reperfusion Injury/chemically induced , Reperfusion Injury/pathology , Sirolimus/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...