Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 6 de 6
1.
Lipids Health Dis ; 22(1): 165, 2023 Oct 04.
Article En | MEDLINE | ID: mdl-37794473

BACKGROUND: Non-high-density lipoprotein-cholesterol (non-HDL-C) has been identified as a potential biomarker for metabolic syndrome (MetS). However, its predictive capability for MetS varies among different ethnic groups, necessitating further investigation. This study aimed to assess the role of non-HDL-C in the early diagnosis of MetS in the Iranian population through a longitudinal study with a 10-year follow-up period. METHODS: Our study enrolled 4684 individuals from the MASHAD (Mashhad Stroke and Heart Atherosclerotic Disorder) cohort who were followed for 10 years to examine the association between non-HDL-C and the incidence of MetS. Additionally, the contribution of individual MetS components to the overall burden was evaluated. RESULTS: A total of 1599 subjects developed MetS, while 3085 did not. Non-HDL-C levels ≥ 130 were associated with a 42% higher risk of developing MetS (relative risk (RR), 1.42; 95% confidence interval (CI), 1.25-1.62). Regarding MetS components, elevated waist circumference (WC) showed the strongest association with MetS incidence (RR, 2.32; 95% CI, 1.45-2.9), whereas triglyceride (TG) levels ≥ 150 mg/dL demonstrated the weakest association (RR, 1.23; 95% CI, 1.04-1.46). Additionally, higher HDL-C levels were reported to be 20% protective against the risk of MetS (RR, 0.8; 95% CI, 0.73-0.86). Moreover, fasting blood glucose (FBG) levels ≥ 100 mg/dL were not significantly linked to MetS burden, while systolic blood pressure (BP) levels ≥ 130 mmHg or diastolic BP levels ≥ 85 mmHg increased the risk of MetS incidence (RR, 1.25; 95% CI: 1.11-1.41). CONCLUSIONS: Elevated non-HDL-C and increased WC serve as significant predictors of MetS in Iranians. Strategies targeting non-HDL-C levels and weight loss should be emphasized to mitigate the risk of MetS development.


Metabolic Syndrome , Humans , Follow-Up Studies , Iran/epidemiology , Longitudinal Studies , Cholesterol , Lipoproteins , Risk Factors , Cholesterol, HDL , Triglycerides
2.
Nutr Metab Cardiovasc Dis ; 33(11): 2055-2066, 2023 Nov.
Article En | MEDLINE | ID: mdl-37567791

AIMS: Studies have indicated inconsistent results regarding the association between plasma levels of Lipoprotein(a) [Lp(a)] and coronary artery calcification (CAC). We performed a systematic review and meta-analysis to investigate the association between elevated levels of Lp(a) and risk of CAC in populations free of cardiovascular disease (CVD) symptoms. DATA SYNTHESIS: PubMed, Web of Science, Embase, and Scopus were searched up to July 2022 and the methodological quality was assessed using Newcastle-Ottawa Scale (NOS) scale. Random-effects meta-analysis was used to estimate pooled odds ratio (OR) and 95% confidence interval. Out of 298 studies, data from 8 cross-sectional (n = 18,668) and 4 cohort (n = 15,355) studies were used in meta-analysis. Cohort studies demonstrated a positive significant association between Lp(a) and CAC, so that individuals with Lp(a)≥30-50 exposed to about 60% risk of CAC incidence compared to those with lower Lp(a) concentrations in asymptomatic CVD subjects (OR, 1.58; 95% CI, 1.38-1.80; l2, 0.0%; P, 0.483); Subgroup analysis showed that a cut-off level for Lp(a) measurement could not statistically affect the association, but race significantly affected the relationship between Lp(a) and CAC (OR,1.60; 95% CI, 1.41-1.81). Analyses also revealed that both men and women with higher Lp(a) concentrations are at the same risk for increased CAC. CONCLUSIONS: Blood Lp(a) level was significantly associated with CAC incidence in asymptomatic populations with CVD, indicating that measuring Lp(a) may be a useful biomarker for diagnosing subclinical atherosclerosis in individuals at higher risk of CAC score. PROSPERO REGISTRATION NUMBER: CRD42022350297.

3.
BMC Cardiovasc Disord ; 23(1): 96, 2023 02 21.
Article En | MEDLINE | ID: mdl-36809976

Coronary artery calcification (CAC) is one of the critical cardiovascular complications that lead to elevated morbidity and mortality among patients with type 2 diabetes (T2M). The association between osteoprotegerin (OPG) and CAC could potentially provide a reasonable chance for preventive therapy in type 2 diabetic patients and benefit the rate of mortality. Since measurement of CAC score is relatively expensive and requires radiation exposure, the current systematic review aims to provide clinical evidence for evaluating the prognostic role of OPG in determining CAC risk among subjects with T2M. Web of Science, PubMed, Embase, and Scopus, were investigated until July 2022. We assessed human studies investigating the association of OPG with CAC in type 2 diabetic patients. Quality assessment was performed by Newcastle-Ottawa quality assessment scales (NOS). Out of 459 records, 7 studies remained eligible to be included. Observational studies that provided odds ratio (OR) estimates with 95% confidence intervals (CIs) for the association between OPG and the risk of CAC were analyzed by random-effects model. In order to provide a visual summary of our findings, the estimation of pooled OR from cross-sectional studies was reported as 2.86 [95% CI 1.49-5.49], which is consistent with the findings of the cohort study. Results revealed that the association between OPG and CAC was significant among diabetic patients. OPG is hypothesized to be a potential marker in predicting the presence of high coronary calcium score among subjects with T2M that could be recognized as a novel target for further pharmacological investigations.


Coronary Artery Disease , Diabetes Mellitus, Type 2 , Vascular Calcification , Humans , Diabetes Mellitus, Type 2/complications , Osteoprotegerin , Cohort Studies , Biomarkers , Cross-Sectional Studies , Coronary Artery Disease/complications , Risk Factors
4.
Inflammopharmacology ; 30(6): 2145-2152, 2022 Dec.
Article En | MEDLINE | ID: mdl-35723850

BACKGROUND: Ulcerative colitis (UC) is one of the chronic diseases which is increasing in prevalence and patients suffer from illness flare-ups. UC standard regimen treatment has various side effects besides the efficacy, so there is an interest in administering complementary medicine to reduce adverse effects and increase the efficacy, as well. The aim of this study was to evaluate the efficacy and anti-inflammatory effect of Thymus kotschyanus as an additive treatment in a randomized double-blind placebo-controlled trial of UC patients. METHODS: Thirty UC out-patients with mesalazine regimen treatment that fulfilled the inclusion criteria were participated in a 12 week trial and were randomly chosen for the treatment and control group. Fifteen patients were administered a placebo as a control and 15 patients were received Thymus kotschyanus extract by a dose of 0.5 g in a day in the treatment group. Laboratory tests were performed at baseline and week 12. The primary outcome was a reduction in fecal calprotectin as the main intestine inflammatory marker. Likewise, reduction in SCCAI, SIDBQ, and SEO indices were considered as secondary aims. RESULTS: Fecal calprotectin was decreased by 54.74% in the treatment group, as compared with the placebo group at week 12 (p = 0.02). A significant reduction in SCCAI was also shown between the two study groups (p = 0.01). Thymus kotschyanus extract was safe and no severe side effects were reported. CONCLUSION: Administration of Thymus kotschyanus revealed improvement in UC symptoms by the intestinal anti-inflammation effect of the plant and could be suggested as a potential additive treatment in UC patients. The study protocol has been registered under the identification code: IRCT20200406046965N2.


Colitis, Ulcerative , Plant Extracts , Humans , Colitis, Ulcerative/drug therapy , Double-Blind Method , Leukocyte L1 Antigen Complex , Mesalamine/therapeutic use , Plant Extracts/therapeutic use , Treatment Outcome , Thymus Plant/chemistry
5.
Cancer Chemother Pharmacol ; 90(1): 1-6, 2022 07.
Article En | MEDLINE | ID: mdl-35635561

Ifosfamide is one of the chemotherapy regimens which potentially causes neurotoxicity in patients up to 30%. Aprepitant is administered as an anti-emetic agent in chemotherapy and regarding the inhibitory effect on CYP3A4, aprepitant can increase the risk of ifosfamide adverse effects. This study aims to systematically investigate the relation of ifosfamide-induced neurotoxicity and aprepitant or fosaprepitant in chemotherapy cancer patients. Four databases including PubMed, Scopus, Web of Science, and Embase were systematically reviewed without language restriction and hand searching was performed until December 2021. Total 1639 publications were retrieved and nine studies fulfilled the eligibility criteria. For quality assessment, we used Newcastle-Ottawa quality assessment scales (NOS) for retrospective cohort studies and Cochrane Collaboration tool to assess the risk of bias for a randomized controlled trial. Overall, the results of our systematic review indicated a positive enhanced trend between neurotoxicity and concomitant use of ifosfamide and aprepitant or fosaprepitant, but the association was not statistically significant. As indicated by our findings, several studies identified low albumin as a risk factor for ifosfamide-induced encephalopathy. However, further clinical studies with a larger population of patients are required to evaluate the clinical significance of ifosfamide-related neurotoxicity and aprepitant or fosaprepitant.


Ifosfamide , Neurotoxicity Syndromes , Aprepitant , Humans , Ifosfamide/adverse effects , Morpholines/adverse effects , Neurotoxicity Syndromes/drug therapy , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/prevention & control , Randomized Controlled Trials as Topic , Retrospective Studies
6.
J Cell Biochem ; 120(5): 6860-6867, 2019 May.
Article En | MEDLINE | ID: mdl-30378159

Breast cancer is one of the most prevalent malignancies among women around the world. RAS proteins require posttranslational modifications, including protein prenylation for proper membrane localization and signaling. Regulation of RAS signaling via specific and novel pharmacological inhibitors is a potentially novel therapeutic approach in breast cancer therapy. This review summarizes the recent knowledge about the clinical value of RAS prenylation pharmacological inhibitors in breast cancer treatment.

...