Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Gen Hosp Psychiatry ; 89: 75-83, 2024.
Article in English | MEDLINE | ID: mdl-38824832

ABSTRACT

BACKGROUND: Evidence suggests that inflammatory processes play a role in the pathophysiology of schizophrenia. Statins exert anti-inflammatory and antioxidant effects and may be effective in improving the symptoms of schizophrenia. This study explored whether statins, as an adjunctive therapy, can alleviate the symptoms of schizophrenia. METHODS: PubMed, EMBASE, and the Cochrane Library were searched for articles published up to March 2023. The risk-of-bias tool for randomized trials was used to assess study quality. Two researchers independently assessed the risks of bias and extracted data. Pooled data on Positive and Negative Syndrome Scale (PANSS) scores were analyzed. A random-effects model was employed to calculate pooled effect sizes. Statistical heterogeneity across studies was assessed using the I2 statistic. All analyses were performed using RevMan5 and Comprehensive Meta-Analysis software. RESULTS: Nine trials enrolling 533 patients in total were included. Add-on statin therapy was found to be associated with a significantly better total PANSS score [standardized mean difference (SMD) = -0.42, 95% confidence interval (CI) -0.75 to -0.09, I2 = 72%; P = 0.01] and PANSS negative subscale score (SMD = -0.26, 95% CI -0.45 to -0.07, I2 = 0%; P = 0.009) in comparison with placebo. However, add-on statin therapy did not appear to improve scores for the PANSS positive and general subscales at the study-defined endpoint (6-24 weeks). CONCLUSIONS: Our meta-analysis indicates that adjunctive statin therapy may confer benefits in ameliorating PANSS negative and total scores. It needs more solid data to confirm the results are related to clinical improvement and functioning.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors , Schizophrenia , Humans , Schizophrenia/drug therapy , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Outcome Assessment, Health Care/statistics & numerical data , Drug Therapy, Combination , Antipsychotic Agents/administration & dosage , Antipsychotic Agents/pharmacology
2.
Thorac Cancer ; 12(21): 2873-2885, 2021 11.
Article in English | MEDLINE | ID: mdl-34545685

ABSTRACT

OBJECTIVE: Currently, several immune checkpoint inhibitors (ICIs) treatment for advanced non-small-cell lung cancer (NSCLC) have been investigated; their overall efficacy and safety remain unclear. METHODS: We searched electronic databases such as PubMed, EMBASE, and the Cochrane library. The randomized controlled trials (RCTs) that compared ICIs with or without chemotherapy to chemotherapy in advanced NSCLC. We collected and compaired thier parameters, including overall survival (OS), progression-free survival (PFS), objective response rate (ORR), and treatment-related adverse events (TRAEs) of grade ≥3. RESULTS: A total of 15 RCTs involving 8869 patients with NSCLC were included. Pembrolizumab plus platinum-based chemotherapy had higher OS and PFS than platinum-based chemotherapy (hazard ratio [HR] 0.55, 95% CI 0.46-0.67; HR 0.54, 95% CI 0.41-0.70, respectively). Pembrolizumab plus platinum-based chemotherapy had higher ranked ORR than platinum-based chemotherapy (odds ratio [OR] 2.92, 95% CI 1.99-4.22). In terms of OS, atezolizumab, pembrolizumab plus platinum-based chemotherapy, and nivolumab plus ipilimumab ranked as the best treatments for patients with programmed death-ligand 1 (PD-L1) expression levels of ≥50%, 1-49%, and <1%, respectively. In terms of PFS, pembrolizumab plus platinum-based chemotherapy ranked as the best treatment for patients with any PD-L1 expression levels. However, ipilimumab plus platinum-based chemotherapy, nivolumab plus platinum-based chemotherapy, and atezolizumab plus platinum-based chemotherapy have higher TRAEs of grade ≥3 than platinum-based chemotherapy. CONCLUSIONS: Pembrolizumab plus platinum-based chemotherapy prevailed in rank in OS, PFS, and ORR benefit. The TRAEs of pembrolizumab plus platinum-based chemotherapy were more than ICI monotherapy and chemotherapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Immune Checkpoint Inhibitors/therapeutic use , Lung Neoplasms/drug therapy , Humans , Progression-Free Survival , Randomized Controlled Trials as Topic
4.
Support Care Cancer ; 29(8): 4885-4892, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33598734

ABSTRACT

OBJECTIVE: The aim of this meta-analysis was to evaluate the effectiveness of glutamine for preventing or treating moderate-to-severe oral mucositis induced by chemotherapy or radiation therapy in patients with cancer. METHODS: PubMed, Cochrane Library, and Embase were searched for eligible randomized controlled trials (RCTs) up to June 2020. The outcomes analyzed were oral mucositis (at all levels of severity). Data were pooled using the random-effects model and are expressed as risk ratios (RRs) and corresponding 95% confidence intervals (CIs). Heterogeneity was assessed and quantified using I2. RESULTS: Sixteen RCTs were included in this review. In this meta-analysis, compared with placebo, glutamine significantly reduced the incidence of grade 3 and 4 oral mucositis induced by chemotherapy or radiation therapy (RR, 0.53; 95% CI, 0.32-0.88). In subgroup analysis, oral glutamine administration (RR, 0.56; 95% CI, 0.34-0.92) and a medium or low daily dose of glutamine (RR, 0.58; 95% CI, 0.44-0.77; RR, 0.53; 95% CI, 0.28-0.94; respectively) decreased risk. Glutamine caused a borderline significant reduction in the risk of grade 3 and 4 oral mucositis induced by radiotherapy (RR, 0.75; 95% CI, 0.58-0.99) and especially in its prevention (RR, 0.51; 95% CI, 0.28-0.94). CONCLUSIONS: Glutamine significantly reduces the risk of oral mucositis during chemotherapy or radiation therapy. Furthermore, large prospective trials are required to support these findings.


Subject(s)
Glutamine/therapeutic use , Neoplasms/complications , Stomatitis/drug therapy , Glutamine/pharmacology , Humans , Prospective Studies , Randomized Controlled Trials as Topic
5.
J Cell Physiol ; 230(9): 2038-48, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25536014

ABSTRACT

Phosphodiesterase (PDE) inhibitors have been suggested as a possible candidate for the treatment of osteopenia, including osteoporosis. KMUP-1 is a novel xanthine derivative with inhibitory activities on the PDE 3, 4, and 5 iso-enzymes to suppress the degradation of cAMP and cGMP. This study aimed to investigate the effect of KMUP-1 on osteoblast differentiation and the underlying cellular and molecular mechanisms. Primary osteoblasts and osteoblastic MC3T3-E1 cells were examined. KMUP-1 enhanced alkaline phosphatase (ALP) activity and mineralization compared to untreated controls in primary osteoblasts and MC3T3-E1 cells. KMUP-1 also increased the mRNA expression of the osteoblastic differentiation markers, including collagen type 1a, ALP, osteocalcin, osteoprotegerin, BMP-2, and Runx2, a key transcription regulator for osteoblastic differentiation. The osteogenic effect of KMUP-1 was abolished by BMP signaling inhibitor, noggin. Furthermore, we found that KMUP-1 upregulated Smad1/5/8 phosphorylations with subsequent BRE-Luc activation confirmed by transient transfection assay. In addition, KMUP-1 inactivated glycogen synthase kinase-3ß (GSK-3ß), with associated nuclear translocation of ß-catenin. Co-treatment with H89 and KT5823, cAMP and cGMP pathway inhibitors, respectively, reversed the KMUP-1-induced activations of Smad1/5/8, ß-catenin, and Runx2. The findings demonstrate for the first time that KMUP-1 can promote osteoblast maturation and differentiation in vitro via BMP-2/Smad1/5/8 and Wnt/ß-catenin pathways. These effects are mediated, in part, by the cAMP and cGMP signaling. Thus, KMUP-1 may be a novel osteoblast activator and a potential new therapy for osteoporosis.


Subject(s)
Bone Morphogenetic Protein 2/biosynthesis , Cell Differentiation/drug effects , Piperidines/administration & dosage , Smad1 Protein/biosynthesis , Smad5 Protein/biosynthesis , Smad8 Protein/biosynthesis , Xanthines/administration & dosage , Animals , Bone Morphogenetic Protein 2/genetics , Calcification, Physiologic , Cell Line , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Gene Expression Regulation, Developmental/drug effects , Mice , Osteoblasts/drug effects , RNA, Messenger/biosynthesis , Smad1 Protein/genetics , Smad5 Protein/genetics , Smad8 Protein/genetics , Wnt Signaling Pathway
6.
Article in English | MEDLINE | ID: mdl-21785641

ABSTRACT

San-Huang-Xie-Xin-Tang (SHXT) is a traditional Chinese medication consisting of three herbs, namely Coptidis rhizome, Scutellariae radix and Rhei rhizome. This study aimed to examine the cardioprotective effects of SHXT in a rat model of acute myocardial apoptosis induced by ischemia/reperfusion (I/R). Vehicle (intravenous saline) or SHXT (intravenous or oral) was administered prior to I/R (occlusion of left coronary artery for 45 min followed by reperfusion for 2 h). In the vehicle group, myocardial I/R caused myocardial infarction with increased plasma cardiac enzymes, severe arrhythmia and mortality. Myocardial apoptosis was induced by I/R as evidenced by DNA ladder and Bcl-2/Bax ratio. In the SHXT group, we found that SHXT significantly reduced plasma levels of cardiac enzymes, arrhythmia scores (from 5 ± 1 to 2 ± 1, P < .01) and mortality rate (from 53 to 0%, P < .01). In addition, pretreatment with intravenous SHXT reduced the infarct size dose-dependently when compared with the vehicle group (10 mg kg(-1): 14.0 ± 0.2 versus 44.5 ± 5.0%, and 30 mg kg(-1): 6.2 ± 1.2% versus 44.5 ± 5.0%, both P < .01). Similarly, oral administration of SHXT reduced the infarct size dose-dependently. Furthermore, SHXT markedly decreased the apoptosis induced by I/R with increased Bcl-2/Bax ratio. Finally, we found that SHXT counteracted the I/R-induced downstream signaling, resulting in increased myocardial eNOS expression and plasma nitrite, and decreased activation of ERK1/2, p38 and JNK. These data suggest that SHXT has cardioprotective effects against I/R-induced apoptosis, and that these effects are mediated, at least in part, by eNOS and MAPK pathways.

7.
Toxicology ; 256(1-2): 13-24, 2009 Feb 04.
Article in English | MEDLINE | ID: mdl-19056458

ABSTRACT

In this research, we conducted an in vitro analysis to evaluate the prostate cancer cells response to labedipinedilol-A in order to determine the effect of this selective alpha(1)-adrenoceptor antagonist to suppress prostate cancer cell growth by affecting cell proliferation and apoptosis. Here, we report that treatment of androgen-sensitive (LNCaP) and androgen-insensitive (PC-3) prostate cancer cells with labedipinedilol-A inhibited cell proliferation in concentration-dependent and time-dependent manners. Moreover, norepinephrine-stimulated proliferation of both cell lines are markedly inhibited by labedipinedilol-A. The probable involvement of alpha(1)-adrenoceptors in this cellular response is suggested. Labedipinedilol-A-induced growth inhibition was associated with G(0)/G(1) arrest, and G(2)/M arrest depending upon concentrations. Cell cycle blockade was associated with reduced amounts of cyclin D1/2, cyclin E, Cdk2, Cdk4, and Cdk6 and increased levels of the Cdk inhibitory proteins (Cip1/p21 and Kip1/p27). In addition, labedipinedilol-A also induced apoptosis in PC-3 cells, as determined by using Hoechst 33342 staining, DNA fragmentation, and Annexin V staining assay. Furthermore, labedipinedilol-A triggered the mitochondrial apoptotic pathway, as indicated by increasing the expression of Bax, but decreasing the level of Bcl-2, resulting in mitochondrial membrane potential loss, cytochrome c release, and activation of caspase-9 and -3. We further investigated the role of MAPK cascades in the anti-proliferative and apoptosis effects of labedipinedilol-A, and confirmed that labedipinedilol-A could activate JNK1/2 but not p38 in both cell lines. Unlike JNK1/2, however, labedipinedilol-A treatment resulted in down-regulation of phospho-ERK1/2 expression. We concluded that labedipinedilol-A possessed the growth-suppressive and apoptotic effects on LNCaP and PC-3 cells by its alpha(1)-adrenoceptor blockade, and the apoptotic effects of labedipinedilol-A primarily through caspases and MAPKs mediated pathways.


Subject(s)
Adrenergic alpha-1 Receptor Antagonists , Adrenergic alpha-Antagonists/pharmacology , Anisoles/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Proliferation/drug effects , Dihydropyridines/pharmacology , Prostatic Neoplasms/pathology , Benzimidazoles , Blotting, Western , Caspase 3/metabolism , Caspase 9/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Fluorescent Dyes , Humans , Male , Membrane Potentials/drug effects , Mitochondrial Membranes/metabolism , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Oncogene Protein v-akt/genetics , Oncogene Protein v-akt/metabolism , Receptors, Adrenergic, alpha-1/metabolism , Signal Transduction/drug effects
8.
Epilepsy Behav ; 11(3): 303-9, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17826356

ABSTRACT

An enriched environment can enhance brain recovery in animals with early-life status epilepticus (SE). The purpose of this study was to determine the effects of early-life SE on spatial memory and hippocampal extracellular signal-regulated kinase (ERK) level, and the possible therapeutic effects of the enriched environment. Rats were assigned randomly to four groups: (1) control rats (nonenriched control); (2) control rats housed in an enriched environment from Postnatal Day (P) 25 to P40 (enriched control); (3) rats in which SE was induced with lithium-pilocarpine (Li-PC) at P21 (nonenriched SE); and (4) rats in which SE was induced with Li-PC at P21 and then housed in an enriched environment from P25 to P40 (enriched SE). As adults, the rats underwent spatial learning and memory tests in the Morris water maze between P50 and P55. At P55, subsets of animals were evaluated for expression of hippocampal ERK1/2 phosphorylation immediately following completion of the Morris water maze. At ~P100, another set of animals was tested for seizure threshold. When studied as adults, only the nonenriched SE group had a spatial memory deficit. The nonenriched SE group also exhibited lower levels of phosphorylated ERK2 as compared with the nonenriched control, enriched control, and enriched SE groups. Both the nonenriched SE and enriched SE groups had reduced seizure thresholds as compared with the nonenriched control and enriched control groups. Results from this study demonstrate that an enriched environment improves spatial memory in rats subjected to early-life SE, possibly through upregulation of phosphorylated ERK2 in the hippocampus. However, an enriched environment has no effect on seizure threshold.


Subject(s)
Cognition/physiology , Environment , Mitogen-Activated Protein Kinase 1/metabolism , Status Epilepticus/physiopathology , Analysis of Variance , Animals , Animals, Newborn , Behavior, Animal/drug effects , Disease Models, Animal , Hippocampus/enzymology , Lithium Chloride , Male , Maze Learning/drug effects , Maze Learning/physiology , Phosphorylation/drug effects , Pilocarpine , Rats , Rats, Sprague-Dawley , Spatial Behavior/drug effects , Status Epilepticus/chemically induced , Status Epilepticus/pathology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...