Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 391
Filter
Add more filters

Publication year range
1.
Virol J ; 21(1): 150, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38965549

ABSTRACT

Porcine reproductive and respiratory syndrome (PRRS) is endemic worldwide, seriously affecting the development of the pig industry, but vaccines have limited protective effects against PRRSV transmission. The aim of this study was to identify potential anti-PRRSV drugs. We examined the cytotoxicity of seven compounds formulated based on the mass ratio of glycyrrhizic acid to matrine and calculated their inhibition rates against PRRSV in vitro. The results showed that the seven compounds all had direct killing and therapeutic effects on PRRSV, and the compounds inhibited PRRSV replication in a time- and dose-dependent manner. The compound with the strongest anti-PRRSV effect was selected for subsequent in vivo experiments. Pigs were divided into a control group and a medication group for the in vivo evaluation. The results showed that pigs treated with the 4:1 compound had 100% morbidity after PRRSV challenge, and the mortality rate reached 75% on the 8th day of the virus challenge. These results suggest that this compound has no practical anti-PRRSV effect in vivo and can actually accelerate the death of infected pigs. Next, we further analyzed the pigs that exhibited semiprotective effects following vaccination with the compound to determine whether the compound can synergize with the vaccine in vivo. The results indicated that pigs treated with the compound had higher mortality rates and more severe clinical reactions after PRRSV infection (p < 0.05). The levels of proinflammatory cytokines (IL-6, IL-8, IL-1ß, IFN-γ, and TNF-α) were significantly greater in the compound-treated pigs than in the positive control-treated pigs (p < 0.05), and there was no synergistic enhancement with the live attenuated PRRSV vaccine (p < 0.05). The compound enhanced the inflammatory response, prompted the body to produce excessive levels of inflammatory cytokines and caused body damage, preventing a therapeutic effect. In conclusion, the present study revealed that the in vitro effectiveness of these agents does not indicate that they are effective in vivo or useful for developing anti-PRRSV drugs. Our findings also showed that, to identify effective anti-PRRSV drugs, comprehensive drug screening is needed, for compounds with solid anti-inflammatory effects both in vitro and in vivo. Our study may aid in the development of new anti-PRRSV drugs.


Subject(s)
Alkaloids , Antiviral Agents , Glycyrrhizic Acid , Matrines , Porcine Reproductive and Respiratory Syndrome , Porcine respiratory and reproductive syndrome virus , Quinolizines , Virus Replication , Animals , Porcine respiratory and reproductive syndrome virus/drug effects , Alkaloids/pharmacology , Quinolizines/pharmacology , Quinolizines/therapeutic use , Swine , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Glycyrrhizic Acid/pharmacology , Glycyrrhizic Acid/therapeutic use , Porcine Reproductive and Respiratory Syndrome/drug therapy , Porcine Reproductive and Respiratory Syndrome/virology , Porcine Reproductive and Respiratory Syndrome/prevention & control , Virus Replication/drug effects , Cytokines/metabolism , Survival Analysis
2.
Drug Dev Res ; 85(4): e22219, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38845211

ABSTRACT

Sepsis is a life-threatening organ dysfunction that endangers patient lives and is caused by an imbalance in the host defense against infection. Sepsis continues to be a significant cause of morbidity and mortality in critically sick patients. Oxymatrine (OMT), a quinolizidine alkaloid derived from the traditional Chinese herb Sophora flavescens Aiton, has been shown to have anti-inflammatory effects on a number of inflammatory illnesses according to research. In this study, we aimed to evaluate the therapeutic effects of OMT on sepsis and explore the underlying mechanisms. We differentiated THP-1 cells into THP-1 macrophages and studied the anti-inflammatory mechanism of OMT in a lipopolysaccharide (LPS)-induced THP-1 macrophage sepsis model. Activation of the receptor for advanced glycation end products (RAGE), as well as NF-κB, was assessed by Western blot analysis and immunofluorescence staining. ELISA was used to measure the levels of inflammatory factors. We found that OMT significantly inhibited HMGB1-mediated RAGE/NF-κB activation and downstream inflammatory cytokine production in response to LPS stimulation. Finally, an in vivo experiment was performed on septic mice to further study the effect of OMT on injured organs. The animal experiments showed that OMT significantly inhibited HMGB1-mediated RAGE/NF-κB activation, protected against the inflammatory response and organ injury induced by CLP, and prolonged the survival rate of septic mice. Herein, we provide evidence that OMT exerts a significant therapeutic effect on sepsis by inhibiting the HMGB1/RAGE/NF-κB signaling pathway.


Subject(s)
Alkaloids , HMGB1 Protein , Inflammation , Lipopolysaccharides , NF-kappa B , Quinolizines , Receptor for Advanced Glycation End Products , Sepsis , Signal Transduction , Alkaloids/pharmacology , Alkaloids/therapeutic use , Quinolizines/pharmacology , Quinolizines/therapeutic use , Animals , Sepsis/drug therapy , Sepsis/complications , Sepsis/metabolism , NF-kappa B/metabolism , HMGB1 Protein/metabolism , HMGB1 Protein/antagonists & inhibitors , Humans , Receptor for Advanced Glycation End Products/metabolism , Signal Transduction/drug effects , Mice , Inflammation/drug therapy , Inflammation/metabolism , Male , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , THP-1 Cells , Mice, Inbred C57BL , Macrophages/drug effects , Macrophages/metabolism , Matrines
3.
Int J Mol Sci ; 25(12)2024 Jun 16.
Article in English | MEDLINE | ID: mdl-38928319

ABSTRACT

Matrine (MT) possesses anti-inflammatory, anti-allergic and antioxidative properties. However, the impact and underlying mechanisms of matrine on colitis are unclear. The purpose of this research was to examine the protective impact and regulatory mechanism of matrine on dextran sulfate sodium (DSS)-induced ulcerative colitis (UC) in mice. MT alleviated DSS-induced UC by inhibiting weight loss, relieving colon shortening and reducing the disease activity index (DAI). Moreover, DSS-induced intestinal injury and the number of goblet cells were reversed by MT, as were alterations in the expression of zonula occludens-1 (ZO-1) and occludin in colon. Simultaneously, matrine not only effectively restored DSS-induced oxidative stress in colonic tissues but also reduced the production of inflammatory cytokines. Furthermore, MT could treat colitis mice by regulating the regulatory T cell (Treg)/T helper 17 (Th17) cell imbalance. We observed further evidence that MT alleviated the decrease in intestinal flora diversity, reduced the proportion of Firmicutes and Bacteroidetes, decreased the proportion of Proteobacteria and increased the relative abundance of Lactobacillus and Akkermansia in colitis mice. In conclusion, these results suggest that MT may mitigate DSS-induced colitis by enhancing the colon barrier integrity, reducing the Treg/Th17 cell imbalance, inhibiting intestinal inflammation, modulating oxidative stress and regulating the gut microbiota. These findings provide strong evidence for the development and application of MT as a dietary treatment for UC.


Subject(s)
Alkaloids , Dextran Sulfate , Gastrointestinal Microbiome , Matrines , Oxidative Stress , Quinolizines , T-Lymphocytes, Regulatory , Animals , Alkaloids/pharmacology , Gastrointestinal Microbiome/drug effects , Oxidative Stress/drug effects , Quinolizines/pharmacology , Quinolizines/therapeutic use , Mice , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Male , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Colitis/microbiology , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Zonula Occludens-1 Protein/metabolism , Colon/pathology , Colon/metabolism , Colon/drug effects , Colon/microbiology , Th17 Cells/drug effects , Th17 Cells/metabolism , Th17 Cells/immunology , Disease Models, Animal , Cytokines/metabolism , Mice, Inbred C57BL , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/pathology , Occludin/metabolism
4.
Brief Bioinform ; 22(5)2021 09 02.
Article in English | MEDLINE | ID: mdl-33876189

ABSTRACT

Targeting tumor microenvironment (TME), such as immune checkpoint blockade (ICB), has achieved increased overall response rates in many advanced cancers, such as non-small cell lung cancer (NSCLC), however, only in a fraction of patients. To improve the overall and durable response rates, combining other therapeutics, such as natural products, with ICB therapy is under investigation. Unfortunately, due to the lack of systematic methods to characterize the relationship between TME and ICB, development of rational immune-combination therapy is a critical challenge. Here, we proposed a systems pharmacology strategy to identify resistance regulators of PD-1/PD-L1 blockade and develop its combinatorial drug by integrating multidimensional omics and pharmacological methods. First, a high-resolution TME cell atlas was inferred from bulk sequencing data by referring to a high-resolution single-cell data and was used to predict potential resistance regulators of PD-1/PD-L1 blockade through TME stratification analysis. Second, to explore the drug targeting the resistance regulator, we carried out the large-scale target fishing and the network analysis between multi-target drug and the resistance regulator. Finally, we predicted and verified that oxymatrine significantly enhances the infiltration of CD8+ T cells into TME and is a powerful combination agent to enhance the therapeutic effect of anti-PD-L1 in a mouse model of lung adenocarcinoma. Overall, the systems pharmacology strategy offers a paradigm to identify combinatorial drugs for ICB therapy with a systems biology perspective of drug-target-pathway-TME phenotype-ICB combination.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/drug effects , Immune Checkpoint Inhibitors/therapeutic use , Lung Neoplasms/drug therapy , Plant Extracts/therapeutic use , Alkaloids/pharmacology , Alkaloids/therapeutic use , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Drug Therapy, Combination , Female , Gene Regulatory Networks/drug effects , Gene Regulatory Networks/genetics , Humans , Immune Checkpoint Inhibitors/pharmacology , Kaplan-Meier Estimate , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice, Inbred C57BL , Plant Extracts/pharmacology , Quinolizines/pharmacology , Quinolizines/therapeutic use , Sophora/chemistry , Treatment Outcome , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics
5.
Nicotine Tob Res ; 25(9): 1547-1555, 2023 08 19.
Article in English | MEDLINE | ID: mdl-37291049

ABSTRACT

INTRODUCTION: A smoking-cessation program was implemented as a randomized non-inferiority trial in primary care practices in Croatia and Slovenia to investigate whether a standard 4-week treatment with cytisine was at least as effective and feasible as a standard 12-week treatment with varenicline in helping smokers quit. AIMS AND METHODS: Out of 982 surveyed smokers, 377 were recruited to the non-inferiority trial: 186 were randomly assigned to cytisine and 191 to varenicline treatment. The primary cessation outcome was 7-day abstinence after 24 weeks, while the primary feasibility outcome was defined by adherence to the treatment plan. We also compared the rates of adverse events between the two treatment groups. RESULTS: The cessation rate after 24 weeks was 32.46% (62/191) in the varenicline group and 23.12% (43/186) in the cytisine group (odds ratio [OR]: 95%, credible interval [CI]: 0.39 to 0.98). Of 191 participants assigned to varenicline treatment 59.16% (113) were adherent, while 70.43% (131 of 186) were adherent in the cytisine group (OR: 1.65, 95% CI: 1.07 to 2.56). Participants assigned to cytisine experienced fewer total (incidence rate ratio [IRR]: 0.59, 95% CI: 0.43 to 0.81) and fewer severe or more extreme adverse events (IRR: 0.72, 95% CI: 0.35 to 1.47). CONCLUSIONS: This randomized non-inferiority trial (n = 377) found the standard 4-week cytisine treatment to be less effective than the standard 12-week varenicline treatment for smoking cessation. However, adherence to the treatment plan, ie, feasibility, was higher, and the rate of adverse events was lower among participants assigned to cytisine treatment. IMPLICATIONS: The present study found the standard 12 weeks of varenicline treatment to be more effective than the standard 4 weeks of cytisine treatment for smoking cessation in a primary care setting in Croatia and Slovenia. Participants assigned to cytisine, however, had a higher adherence to the treatment plan and a lower rate of adverse events. Estimates from the present study may be especially suitable for generalizations to high-smoking prevalence populations in Europe. Given the much lower cost of cytisine treatment, its lower rate of adverse events, and higher feasibility (but its likely lower effectiveness with the standard dosage regimen), future analyses should assess the cost-effectiveness of the two treatments for health policy considerations.


Subject(s)
Alkaloids , Smoking Cessation , Humans , Alkaloids/therapeutic use , Azocines/therapeutic use , Benzazepines/adverse effects , Nicotine/adverse effects , Nicotinic Agonists/adverse effects , Primary Health Care , Quinolizines/therapeutic use , Treatment Outcome , Varenicline/therapeutic use
6.
Neurochem Res ; 47(5): 1369-1382, 2022 May.
Article in English | MEDLINE | ID: mdl-35099719

ABSTRACT

Hyperglycemia aggravates cerebral ischemia/reperfusion (I/R) injury via vascular injury. There is still a lack of effective pharmaceutical preparations for cerebral I/R injury under hyperglycemia. This study aimed to investigate the effects of oxymatrine (OMT) on hyperglycemia-exacerbated cerebral I/R injury in vitro and in vivo. The middle cerebral artery occlusion (MCAO) and reperfusion was established in the rats under hyperglycemia. Meanwhile, oxygen-glucose deprivation and reoxygenation (OGD/R) with high glucose was used as an in vitro model of hyperglycemic cerebral I/R injury. The results showed that the neurological deficit score, mortality, infarct volume and penumbra apoptosis in hyperglycemia group were significantly higher than those in normal glucose group. OMT pre-treated obviously reduced the degree of neurological deficit, mortality, infarct volume, improve cerebral blood flow after I/R in rats with hyperglycemia, and increase the survival rate of human brain microvascular endothelial cells (HBMECs) in high glucose and OGD/R group. OMT significantly improved the ultrastructure changes of endothelial cells, and maintain the migration and angiogenesis potency of HBMECs in high glucose and OGD/R group. OMT obviously alleviated the down-regulating CD31 and CD105 expression in cerebral microvessels caused by hyperglycemia. It is concluded that OMT treatment might alleviate cerebral I/R injury under hyperglycemia via protecting microvessels.


Subject(s)
Alkaloids , Brain Ischemia , Quinolizines , Reperfusion Injury , Alkaloids/therapeutic use , Animals , Apoptosis , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Endothelial Cells/metabolism , Humans , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/metabolism , Microvessels/metabolism , Quinolizines/therapeutic use , Rats , Rats, Sprague-Dawley , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism
7.
J Appl Toxicol ; 42(8): 1354-1370, 2022 08.
Article in English | MEDLINE | ID: mdl-35146781

ABSTRACT

Fluoroquinolone (FQ) antibacterials have drawn heightened attention from various international regulatory agencies due to their class-specific side effects. Levonadifloxacin is a novel broad spectrum benzoquinolizine FQ active against methicillin-resistant Staphyloccocus aureus (MRSA). Owing to FQ-associated safety concerns, extensive preclinical safety pharmacology (central nervous system and cardiac safety) and toxicology studies (subacute repeat-dose toxicity, genotoxicity, phototoxicity and chondrotoxicity) of levonadifloxacin were performed at relatively high doses. Intravenous (IV) and oral studies were conducted using WCK 771 (l-arginine salt of levonadifloxacin) and WCK 2349 (l-alanine ester prodrug of levonadifloxacin), respectively. Safety pharmacology studies following single dose revealed no adverse effects on central nervous system (including seizure) in mice and cardiovascular system (hERG and monkey telemetry). In repeat-dose toxicity studies, except for IV bolus dosing related effects in rat (hyperactivity, mild convulsion, polypnoea and injection site irritation) and dog (emesis and salivation), no other adverse findings limiting the dosing duration were observed. No major biochemical, haematological, gross or histopathological changes suggestive of damage to vital organs were observed in either WCK 771- or WCK 2349-treated groups. WCK 771 and WCK 2349 were found to be nongenotoxic; however, they showed weak phototoxicity that was comparable with levofloxacin. WCK 771 showed chondrotoxicity in the Beagle dog pups on repeat-dose administration; however, the severity level was lower than ofloxacin. Overall, preclinical safety studies helped establish wider safety margin for WCK 771 and WCK 2349 that supports administration of higher therapeutic doses in humans by both IV and oral routes, thereby enabling safe anti-MRSA treatment.


Subject(s)
Fluoroquinolones , Quinolones , Administration, Oral , Animals , Anti-Bacterial Agents/toxicity , Dogs , Humans , Mice , Quinolizines/pharmacology , Quinolizines/therapeutic use , Rats
8.
J Cell Mol Med ; 25(1): 549-560, 2021 01.
Article in English | MEDLINE | ID: mdl-33210432

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most lethal cancer types with insufficient approved therapies, among which lenvatinib is a newly approved multi-targeted tyrosine kinase inhibitor for frontline advanced HCC treatment. However, resistance to lenvatinib has been reported in HCC treatment recently, which limits the clinical benefits of lenvatinib. This study aims to investigate the underlying mechanism of lenvatinib resistance and explore the potential drug to improve the treatment for lenvatinib-resistant (LR) HCC. Here, we developed two human LR HCC cell lines by culturing with long-term exposure to lenvatinib. Results showed that the vascular endothelial growth factor receptors (VEGFR)2 expression and its downstream RAS/MEK/ERK signalling were obviously up-regulated in LR HCC cells, whereas the expression of VEGFR1, VEGFR3, FGFR1-4 and PDGFRα/ß showed no difference. Furthermore, ETS-1 was identified to be responsible for VEGFR2 mediated lenvatinib resistance. The cell models were further used to explore the potential strategies for restoration of sensitivity of lenvatinib. Sophoridine, an alkaloid extraction, inhibited the proliferation, colony formation, cell migration and increased apoptosis of LR HCC cells. In vivo and in vitro results showed Sophoridine could further sensitize the therapeutic of lenvatinib against LR HCC. Mechanism studies revealed that Sophoridine decreased ETS-1 expression to down-regulate VEGFR2 expression along with downstream RAS/MEK/ERK axis in LR HCC cells. Hence, our study revealed that up-regulated VEGFR2 expression could be a predicator of the resistance of lenvatinib treatment against HCC and provided a potential candidate to restore the sensitivity of lenvatinib for HCC treatment.


Subject(s)
Alkaloids/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Phenylurea Compounds/pharmacology , Quinolines/pharmacology , Quinolizines/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Alkaloids/therapeutic use , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Humans , Phenylurea Compounds/therapeutic use , Quinolines/therapeutic use , Quinolizines/therapeutic use , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-3/genetics , Vascular Endothelial Growth Factor Receptor-3/metabolism , Matrines
9.
Pharmacol Res ; 171: 105778, 2021 09.
Article in English | MEDLINE | ID: mdl-34298110

ABSTRACT

The plant Sophora flavescens Ait. has been used in the clinical management of colorectal cancer (CRC). Its constituent compounds, notably the alkaloids matrine, oxymatrine, and sophoridine, have received considerable research attention in experimental models of CRC in vivo and in vitro. This review found that extracts of S. flavescens and/or its constituent compounds have been reported to inhibit CRC cell proliferation by inducing cell-cycle arrest at the G1 phase, inducing apoptosis via the intrinsic pathway, interfering in cancer metabolism, inhibiting metastasis and angiogenesis, regulating senescence and telomeres, regulating the tumour microenvironment and down-regulating cancer-related inflammation. In addition, matrine and oxymatrine reversed multi-drug resistance and enhanced the effects of chemotherapies. These anti-cancer effects were associated with regulation of several cellular signalling pathways including: MAPK/ERK, PI3K/AKT/mTOR, p38MAPK, NF-κB, Hippo/LATS2, TGF-ß/Smad, JAK/STAT3, RhoA/ROC, and Wnt/ ß-catenin pathways. These multiple actions in CRC suggest the alkaloids of S. flavescens may be therapeutic candidates for CRC management. Nevertheless, there remains considerable scope for future research into its flavonoid constituents, the effects of combinations of compounds, and the interaction between these compounds and anti-cancer drugs. In addition, more research is needed to investigate likely drug ligand-receptor interactions for each of the bioactive compounds.


Subject(s)
Alkaloids/therapeutic use , Antineoplastic Agents, Phytogenic/therapeutic use , Colorectal Neoplasms/drug therapy , Quinolizines/therapeutic use , Sophora , Animals , Humans , Phytotherapy , Matrines
10.
Acta Pharmacol Sin ; 42(4): 573-584, 2021 Apr.
Article in English | MEDLINE | ID: mdl-32694761

ABSTRACT

Pathological cardiac fibrosis is a common feature in multiple cardiovascular diseases that contributes to the occurrence of heart failure and life-threatening arrhythmias. Our previous study demonstrated that matrine could attenuate doxorubicin-induced oxidative stress and cardiomyocyte apoptosis. In this study, we investigated the effect of matrine on cardiac fibrosis. Mice received aortic banding (AB) operation or continuous injection of isoprenaline (ISO) to generate pathological cardiac fibrosis and then were exposed to matrine lavage (200 mg·kg-1·d-1) or an equal volume of vehicle as the control. We found that matrine lavage significantly attenuated AB or ISO-induced fibrotic remodeling and cardiac dysfunction. We also showed that matrine (200 µmol/L) significantly inhibited the proliferation, migration, collagen production, and phenotypic transdifferentiation of cardiac fibroblasts. Mechanistically, matrine suppressed p38 activation in vivo and in vitro, and overexpression of constitutively active p38 completely abolished the protective effects of matrine. We also demonstrated that ribosomal protein S5 (RPS5) upregulation was responsible for matrine-mediated inhibition on p38 and fibrogenesis. More importantly, matrine was capable of ameliorating preexisting cardiac fibrosis in mice. In conclusion, matrine treatment attenuates cardiac fibrosis by regulating RPS5/p38 signaling in mice, and it might be a promising therapeutic agent for treating pathological cardiac fibrosis.


Subject(s)
Alkaloids/therapeutic use , Cardiomyopathies/drug therapy , Cardiotonic Agents/therapeutic use , Fibrosis/drug therapy , Quinolizines/therapeutic use , Ribosomal Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cardiomyopathies/chemically induced , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Transdifferentiation/drug effects , Fibroblasts/drug effects , Fibrosis/chemically induced , Heart/drug effects , Isoproterenol , MAP Kinase Signaling System/drug effects , Male , Mice, Inbred C57BL , Up-Regulation/drug effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Matrines
11.
JAMA ; 326(1): 56-64, 2021 07 06.
Article in English | MEDLINE | ID: mdl-34228066

ABSTRACT

Importance: Cytisine is more effective than placebo and nicotine replacement therapy for smoking cessation. However, cytisine has not been tested against the most effective smoking cessation medication, varenicline, which is associated with adverse events known to lead to discontinuation of therapy. Objective: To examine whether standard cytisine treatment (25 days) was at least as effective as standard varenicline treatment (84 days) for smoking cessation. Design, Setting, and Participants: This noninferiority, open-label randomized clinical trial with allocation concealment and blinded outcome assessment was undertaken in Australia from November 2017 through May 2019; follow-up was completed in January 2020. A total of 1452 Australian adult daily smokers willing to make a quit attempt were included. Data collection was conducted primarily by computer-assisted telephone interview, but there was an in-person visit to validate the primary outcome. Interventions: Treatments were provided in accordance with the manufacturers' recommended dosage: cytisine (n = 725), 1.5-mg capsules taken 6 times daily initially then gradually reduced over the 25-day course; varenicline (n = 727), 0.5-mg tablets titrated to 1 mg twice daily for 84 days (12 weeks). All participants were offered referral to standard telephone behavioral support. Main Outcomes and Measures: The primary outcome was 6-month continuous abstinence verified using a carbon monoxide breath test at 7-month follow-up. The noninferiority margin was set at 5% and the 1-sided significance threshold was set at .025. Results: Among 1452 participants who were randomized (mean [SD] age, 42.9 [12.7] years; 742 [51.1%] women), 1108 (76.3%) completed the trial. Verified 6-month continuous abstinence rates were 11.7% for the cytisine group and 13.3% for the varenicline group (risk difference, -1.62% [1-sided 97.5% CI, -5.02% to ∞]; P = .03 for noninferiority). Self-reported adverse events occurred less frequently in the cytisine group (997 events among 482 participants) compared with the varenicline group (1206 events among 510 participants) and the incident rate ratio was 0.88 (95% CI, 0.81 to 0.95; P = .002). Conclusions and Relevance: Among daily smokers willing to quit, cytisine treatment for 25 days, compared with varenicline treatment for 84 days, failed to demonstrate noninferiority regarding smoking cessation. Trial Registration: anzctr.org.au Identifier: ACTRN12616001654448.


Subject(s)
Alkaloids/therapeutic use , Smoking Cessation Agents/therapeutic use , Smoking Cessation/methods , Varenicline/therapeutic use , Adult , Alkaloids/adverse effects , Azocines/adverse effects , Azocines/therapeutic use , Dreams , Female , Humans , Male , Middle Aged , Nausea/chemically induced , Quinolizines/adverse effects , Quinolizines/therapeutic use , Smoking Cessation Agents/adverse effects , Treatment Outcome , Varenicline/adverse effects
12.
Metab Brain Dis ; 35(1): 149-157, 2020 01.
Article in English | MEDLINE | ID: mdl-31840202

ABSTRACT

The present study was performed to examine the effect of oxymatrine (OMT) on motor functions and histopathologic changes after spinal cord injury and the mechanism underlying its neuroprotective effects. Results suggested that, OMT causes regain of lost motor function near to normal via attenuating oxidative stress, inflammatory response and cellular apoptosis. These observations were further supported by histological examination of spinal cord of rats. It also showed to regulate pro-inflammatory cytokines, Bcl2 family proteins and reduces the level of toll like receptor (TLR-4) and nuclear factor-kappa B (NF-ĸB) in concentration dependent manner. The mitogen-activated protein kinase (MAPK) pathway was also regulated by OMT after SCI. It has been suggested that, OMT promotes the recovery of motor function after SCI in rats via multiple mechanism, and this effect may be related to its anti-oxidant, anti-inflammatory and anti-apoptotic effects.


Subject(s)
Alkaloids/therapeutic use , Inflammation Mediators/metabolism , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Quinolizines/therapeutic use , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/prevention & control , Alkaloids/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Dose-Response Relationship, Drug , Inflammation/metabolism , Inflammation/pathology , Inflammation/prevention & control , Inflammation Mediators/antagonists & inhibitors , Male , Neuroprotective Agents/pharmacology , Oxidative Stress/physiology , Quinolizines/pharmacology , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/pathology
13.
Pharmacol Res ; 147: 104327, 2019 09.
Article in English | MEDLINE | ID: mdl-31283981

ABSTRACT

Oxymatrine (OMT) is a quinolizidine alkaloid derived from the roots of the Sophora genus plants. It has been widely used as a treatment for chronic hepatitis infections and inflammatory diseases due to its effective immunomodulatory and anti-inflammatory properties. Recently, the potential anti-cancer effects of OMT have been actively studied in various cancers. It can induce apoptosis and inhibit the proliferation of tumor cells, including those of colorectal cancer, gall bladder carcinoma, and leukemia. Moreover, it reduces tumor growth in different in vivo models as well as augments the anti-cancer effects of existing chemotherapeutics on tumor cells. OMT regulates various oncogenic signaling pathways such as the Akt, epidermal growth factor receptor (EGFR), and nuclear factor kappa B (NF-κB) cascades to exert its cytotoxicity against cancer cells. This review provides an overview of the current knowledge on the potential of OMT as an anti-cancer therapeutic through the modulation of diverse oncogenic molecular targets.


Subject(s)
Alkaloids/therapeutic use , Antineoplastic Agents, Phytogenic/therapeutic use , Neoplasms/drug therapy , Quinolizines/therapeutic use , Alkaloids/chemistry , Alkaloids/pharmacology , Animals , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Disease Models, Animal , Humans , Quinolizines/chemistry , Quinolizines/pharmacology
14.
Bioorg Chem ; 90: 103032, 2019 09.
Article in English | MEDLINE | ID: mdl-31207450

ABSTRACT

A series of new cytisine derivatives with a unique endocyclic scaffold were synthesized and evaluated for their inhibitory effect on collagen α1 (I) (COL1A1) promotor in human LX2 cells, taking cytisine as the lead. Structure-activity relationship (SAR) revealed that introducing a 12N-benzyl substitution might significantly enhance the activity. Compound 5f exhibited a promising inhibitory potency against COL1A1 with an IC50 value of 12.8 µM in human LX2 cells, and an inspiring inhibition activity against COL1A1 on both mRNA and protein levels. It also effectively inhibited the expression of α smooth muscle actin (α-SMA), connective tissue growth factor (CTGA), matrix metalloprotein 2 (MMP-2), and transforming growth factor ß1 (TGFß1), indicating an extensive inhibitory effect against fibrogenetic proteins. In addition, compound 5f displayed reasonable PK and safety profiles. The primary mechanism study indicated that it might repress the hepatic fibrogenesis via PI3K/Akt/Smad signaling pathway. The results provided powerful information for further structure optimization, and compound 5f was selected as a novel anti-liver fibrosis agent for further investigation.


Subject(s)
Alkaloids/therapeutic use , Liver Cirrhosis/drug therapy , Signal Transduction/drug effects , Alkaloids/chemical synthesis , Alkaloids/pharmacokinetics , Animals , Azocines/chemical synthesis , Azocines/pharmacokinetics , Azocines/therapeutic use , Cell Line , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Humans , Male , Mice , Molecular Structure , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Quinolizines/chemical synthesis , Quinolizines/pharmacokinetics , Quinolizines/therapeutic use , Rats, Sprague-Dawley , Smad Proteins/metabolism , Structure-Activity Relationship
15.
Can J Physiol Pharmacol ; 97(2): 99-106, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30485133

ABSTRACT

Amyloid beta 42 (Aß1-42)-induced oxidative stress causes the death of neuronal cells and is involved in the development of Alzheimer's disease. Oxymatrine (OMT) inhibits oxidative stress. In this study, we investigated the effect of OMT on Aß1-42-induced neurotoxicity in vivo and in vitro. In the Morris water maze test, OMT significantly decreased escape latency and increased the number of platform crossings. In vitro, OMT markedly increased cell viability and superoxide dismutase activity. Moreover, OMT decreased lactate dehydrogenase leakage, malondialdehyde content, and reactive oxygen species in a dose-dependent manner. OMT upregulated the ratio of Bcl-2/Bax and downregulated the level of caspase-3. Furthermore, OMT inhibited the activation of MAP kinase (ERK 1/2, JNK) and nuclear factor κB. In summary, OMT may potentially be used in the treatment of Alzheimer's disease.


Subject(s)
Alkaloids/pharmacology , Alzheimer Disease/drug therapy , Maze Learning/drug effects , Memory/drug effects , Neurons/drug effects , Quinolizines/pharmacology , Alkaloids/therapeutic use , Alzheimer Disease/chemically induced , Alzheimer Disease/pathology , Amyloid beta-Peptides/toxicity , Animals , Cells, Cultured , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Humans , Male , Neurons/pathology , Oxidative Stress/drug effects , Peptide Fragments/toxicity , Primary Cell Culture , Quinolizines/therapeutic use , Rats , Rats, Sprague-Dawley
16.
Pharmacology ; 104(5-6): 235-243, 2019.
Article in English | MEDLINE | ID: mdl-31357205

ABSTRACT

BACKGROUNDS: (6aS, 10S, 11aR, 11bR, 11cS)-10-methylaminododecahydro-3a, 7a-diaza-benzo (de) anthracene-8-thione (MASM), a novel derivative of matrine, exhibits better anti-inflammatory activity. This study was designed to evaluate the protective effect of MASM on acute and chronic liver injuries and explore the possible mechanisms. METHODS: Acute and chronic liver injury models were established by the CCl4 intraperitoneal injection and the protective effect of MASM was assessed by biochemical and histological examination. The infiltration of different monocyte subsets into the liver was characterized and analyzed by flow cytometry. The in vitro effect of MASM on liver nonparenchymal cells was evaluated by real-time PCR and transwell chemotaxis assays. RESULTS: Administration of MASM markedly attenuated acute liver injury and liver fibrosis induced by CCl4 injection. Meanwhile, the infiltrations of Gr1hi monocytes in injured livers and induced hepatic expression of monocyte chemoattractant protein-1 (MCP-1) were greatly inhibited. Cellular experiments demonstrated that MASM not only decreased the expression of MCP-1 but also inhibited its chemotactic activity. CONCLUSIONS: This study demonstrates that the protective effect of MASM on liver injury could be contributed to the suppression of Gr1hi monocyte infiltration to the liver and the inhibition of MCP-1 production and activity. These findings provide new insights into the protective role of MASM in liver injury.


Subject(s)
Alkaloids/therapeutic use , Anthracenes/pharmacology , Anti-Inflammatory Agents/therapeutic use , Chemical and Drug Induced Liver Injury/drug therapy , Liver Cirrhosis/drug therapy , Monocytes/drug effects , Quinolizines/therapeutic use , Thiones/pharmacology , Alkaloids/pharmacology , Animals , Anthracenes/therapeutic use , Anti-Inflammatory Agents/pharmacology , Antigens, Ly/immunology , Carbon Tetrachloride , Chemical and Drug Induced Liver Injury/immunology , Chemical and Drug Induced Liver Injury/pathology , Chemokine CCL2/immunology , Liver/drug effects , Liver/immunology , Liver/pathology , Liver Cirrhosis/immunology , Liver Cirrhosis/pathology , Male , Mice, Inbred C57BL , Monocytes/immunology , Quinolizines/pharmacology , Thiones/therapeutic use , Matrines
17.
Metab Brain Dis ; 34(1): 173-181, 2019 02.
Article in English | MEDLINE | ID: mdl-30406376

ABSTRACT

The present study was designed to evaluate the effects of matrine (MAT) on scopolamine (SCOP)-induced learning and memory impairment. After successive oral administration of MAT to mice for three days at doses of 0.4, 2, and 10 mg/kg, we assessed improvements in learning and memory and investigated the mechanism of action of SCOP-induced amnesia. Donepezil at a dose of 3 mg/kg was used as a standard memory enhancer. MAT significantly improved SCOP-induced learning and memory impairment in novel object recognition and Y-maze tests at doses of 0.4, 2, and 10 mg/kg. Furthermore, MAT inhibited acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) activities and decreased oxidative stress in the brain, as evidenced by increased total antioxidant capacity, total superoxide dismutase levels, and catalase activities as well as decreased malondialdehyde levels. Additionally, there was a significant negative correlation between the percentage of spontaneous alternation in the Y maze and AChE activity in the cortex and hippocampus. MAT ameliorated SCOP-induced amnesia by the inhibition of both AChE/BuChE activities and oxidative stress. This study provides further evidence to encourage the development of MAT as a drug for the prevention or treatment of Alzheimer's disease.


Subject(s)
Alkaloids/therapeutic use , Amnesia/drug therapy , Antioxidants/therapeutic use , Cholinesterase Inhibitors/therapeutic use , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Quinolizines/therapeutic use , Alkaloids/pharmacology , Amnesia/chemically induced , Amnesia/metabolism , Animals , Antioxidants/pharmacology , Brain/drug effects , Brain/metabolism , Catalase/metabolism , Cholinesterase Inhibitors/pharmacology , Male , Malondialdehyde/metabolism , Memory/drug effects , Mice , Mice, Inbred ICR , Neuroprotective Agents/pharmacology , Quinolizines/pharmacology , Scopolamine , Superoxide Dismutase/metabolism , Matrines
18.
Annu Rev Med ; 67: 467-86, 2016.
Article in English | MEDLINE | ID: mdl-26332005

ABSTRACT

The tobacco addiction treatment field is progressing through innovations in medication development, a focus on precision medicine, and application of new technologies for delivering support in real time and over time. This article reviews the evidence for combined and extended cessation pharmacotherapy and behavioral strategies including provider advice, individual counseling, group programs, the national quitline, websites and social media, and incentives. Healthcare policies are changing to offer cessation treatment to the broad population of smokers. With knowledge of the past and present, this review anticipates what is likely on the horizon in the clinical and public health effort to address tobacco addiction.


Subject(s)
Nicotine/administration & dosage , Nicotine/adverse effects , Smoking Cessation/methods , Tobacco Use Cessation Devices , Tobacco Use Disorder/therapy , Alkaloids/therapeutic use , Azocines/therapeutic use , Bupropion/therapeutic use , Directive Counseling , Dopamine Uptake Inhibitors/therapeutic use , Drug Therapy, Combination , Health Policy , Humans , Nicotine/antagonists & inhibitors , Nicotine/metabolism , Nicotinic Agonists/therapeutic use , Quinolizines/therapeutic use , Social Media , Text Messaging , Tobacco Use Disorder/etiology , Varenicline/therapeutic use
19.
Neurochem Res ; 43(8): 1575-1586, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29948728

ABSTRACT

The aim of the study was to elucidate the therapeutic effects of Cytisine (CYT) on cerebral ischemia-reperfusion injury in mice. Male ICR mice were pretreated with reagents (drug), and then subjected to 2 h focal cerebral ischemia and 24 h reperfusion. Morphologically, the histopathological impairment were estimated by the TTC, HE and TUNEL staining. The expression of GluN2B-containing NMDA receptor, phosphorylation of extracellular regulated protein kinases, total ERK, phosphorylation of cAMP-response element binding protein and total CREB were determined by immunofluorescence and Western blot assay, respectively. The mRNA expression of NR2B, ERK and CREB were quantified by the real-time RT-PCR. CYT significantly diminished the infarct size and neuronal apoptosis. Additionally, it ameliorated histopathological lesion dramatically. CYT promoted the phosphorylation of ERK, CREB and their mRNA expression. In contrast, the expression of NR2B was suppressed in concomitant with the down-regulation of genes. The overall results thus far suggest that CYT confers the neuroprotection against cerebral I/R injury by regulating the NR2B-ERK/CREB signal pathway.


Subject(s)
Alkaloids/therapeutic use , Brain Ischemia/prevention & control , Cyclic AMP Response Element-Binding Protein/physiology , MAP Kinase Signaling System/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Reperfusion Injury/prevention & control , Alkaloids/chemistry , Alkaloids/pharmacology , Animals , Azocines/chemistry , Azocines/pharmacology , Azocines/therapeutic use , Brain Ischemia/pathology , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Inbred ICR , Neuroprotection/drug effects , Neuroprotection/physiology , Quinolizines/chemistry , Quinolizines/pharmacology , Quinolizines/therapeutic use , Reperfusion Injury/pathology , Signal Transduction/drug effects , Signal Transduction/physiology
20.
Acta Pharmacol Sin ; 39(11): 1753-1759, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29980742

ABSTRACT

The present study investigated the efficacy of the hepatoprotective drug matrine (Mtr) for its new application for hepatosteatosis and associated disorders in glucose homeostasis. The study was performed in two nutritional models of hepatosteatosis in mice with various abnormal glucose homeostasis: (1) high-fructose diet (HFru) induced hepatosteatosis and glucose intolerance from hepatic, and (2) hepatosteatosis and hyperglycemia induced by high-fat (HF) diet in combination with low doses of streptozotocin (STZ). Administration of Mtr (100 mg/kg every day in diet for 4 weeks) abolished HFru-induced hepatosteatosis and glucose intolerance. These effects were associated with the inhibition of HFru-stimulated de novo lipogenesis (DNL) without altering hepatic fatty acid oxidation. Further investigation revealed that HFru-induced endoplasmic reticulum (ER) stress was inhibited, whereas heat-shock protein 72 (an inducible chaperon protein) was increased by Mtr. In a type 2 diabetic model induced by HF-STZ, Mtr reduced hepatosteatosis and improved attenuated hyperglycemia. The hepatoprotective drug Mtr may be repurposed for the treatment of hepatosteatosis and associated disorders in glucose homeostasis. The inhibition of ER stress associated DNL and fatty acid influx appears to play an important role in these metabolic effects.


Subject(s)
Alkaloids/therapeutic use , Drug Repositioning , Fatty Liver/drug therapy , Glucose Intolerance/drug therapy , Protective Agents/therapeutic use , Quinolizines/therapeutic use , Adiposity/drug effects , Animals , Body Weight/drug effects , Diet, High-Fat/adverse effects , Endoplasmic Reticulum Stress/drug effects , Fatty Acids/metabolism , Fructose/adverse effects , Fructose/metabolism , HSP72 Heat-Shock Proteins/metabolism , Homeostasis/drug effects , Lipogenesis/drug effects , Liver/physiopathology , Mice, Inbred C57BL , Triglycerides/metabolism , Matrines
SELECTION OF CITATIONS
SEARCH DETAIL