Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Lipids Health Dis ; 23(1): 116, 2024 Apr 20.
Article in English | MEDLINE | ID: mdl-38643110

ABSTRACT

BACKGROUND: Insufficient attention has been given to examining the correlation between body composition and hyperuricemia, leading to inconsistent findings. The primary objective of this research is to explore the association between lean body mass index (LMI), visceral fat mass index (VFMI), and hyperuricemia. A specific emphasis will be placed on assessing the link between the ratio of lean body mass to visceral fat mass (LMI/VFMI) and hyperuricemia. METHODS: The present study employed a cross-sectional design and involved a total of 9,646 individuals who participated in the National Health and Nutrition Examination Survey (NHANES). To explore the associations among the variables, logistic and linear regressions were employed. Additionally, subgroup analyses and sensitivity analyses were conducted based on various characteristics. RESULTS: The results showed that LMI was positively associated with hyperuricemia (for Per-SD: OR = 1.88, 95%CI: 1.75, 2.01; for quartiles [Q4:Q1]: OR = 5.37, 95%CI: 4.31, 6.69). Meanwhile, VFMI showed a positive association with hyperuricemia (for Per-SD: OR = 2.02, 95%CI: 1.88, 2.16; for quartiles [Q4:Q1]: OR =8.37, 95%CI: 6.70, 10.47). When considering the effects of In LMI/VFMI, an L-shaped negative association with hyperuricemia was observed (for Per-SD: OR = 0.45, 95%CI: 0.42, 0.49; for quartiles [Q4:Q1]: OR = 0.16, 95%CI: 0.13, 0.20). Subgroup and sensitivity analyses demonstrated the robustness of this association across different subgroups. Additionally, the segmented regression analysis indicated a saturation effect of 5.64 for the In LMI/VFMI with hyperuricemia (OR = 0.20, 95%CI: 0.17, 0.24). For every 2.72-fold increase of In LMI/VFMI, the risk of hyperuricemia was reduced by 80%. CONCLUSION: The LMI/VFMI ratio is non-linearly associated with serum uric acid. Whether this association is causal needs to be confirmed in further longitudinal studies or Mendelian randomization.


Subject(s)
Hyperuricemia , Humans , Cross-Sectional Studies , Nutrition Surveys , Intra-Abdominal Fat , Uric Acid , Body Composition , Body Mass Index
2.
Front Endocrinol (Lausanne) ; 14: 1271857, 2023.
Article in English | MEDLINE | ID: mdl-38089605

ABSTRACT

Background: Insulin resistance (IR), a risk factor for cardiovascular diseases, has garnered significant attention in scientific research. Several studies have investigated the correlation between IR and coronary artery calcification (CAC), yielding varying results. In light of this, we conducted a systematic review to investigate the association between IR as evaluated by the homeostasis model assessment (HOMA-IR) and CAC. Methods: A comprehensive search was conducted to identify relevant studies in PubMed, Embase, Scopus, and Web of Science databases. In addition, preprint servers such as Research Square, BioRxiv, and MedRxiv were manually searched. The collected data were analyzed using either fixed or random effects models, depending on the heterogeneity observed among the studies. The assessment of the body of evidence was performed using the GRADE approach to determine its quality. Results: The current research incorporated 15 studies with 60,649 subjects. The analysis revealed that a higher category of HOMA-IR was associated with a greater prevalence of CAC in comparison to the lowest HOMA-IR category, with an OR of 1.13 (95% CI: 1.06-1.20, I2 = 29%, P < 0.001). A similar result was reached when HOMA-IR was analyzed as a continuous variable (OR: 1.27, 95% CI: 1.14-1.41, I2 = 54%, P < 0.001). In terms of CAC progression, a pooled analysis of two cohort studies disclosed a significant association between increased HOMA-IR levels and CAC progression, with an OR of 1.44 (95% CI: 1.04-2.01, I2 = 21%, P < 0.05). It is important to note that the strength of the evidence was rated as low for the prevalence of CAC and very low for the progression of CAC. Conclusion: There is evidence to suggest that a relatively high HOMA-IR may be linked with an increased prevalence and progression of CAC.


Subject(s)
Coronary Artery Disease , Hyperinsulinism , Insulin Resistance , Humans , Coronary Artery Disease/epidemiology , Coronary Artery Disease/etiology , Risk Factors , Homeostasis
3.
Arch Public Health ; 81(1): 180, 2023 Oct 06.
Article in English | MEDLINE | ID: mdl-37803458

ABSTRACT

BACKGROUND: Studies have explored the correlation between body composition and bone mineral density (BMD), but there has yet to be a consensus. Thus, the present study aims to comprehensively investigate the association between lean body mass, adipose tissue, and BMD. METHODS: We conducted a cross-sectional study using data from the National Health and Nutrition Examination Survey (NHANES) (2011-2018) with 11,227 subjects. Multiple linear regression, smoothed curve fitting, threshold, and saturation effect analysis were used to explore the association between lean body mass, visceral fat mass, and BMD. Also, we used the lean body mass to visceral fat mass ratio (Log LM/VFM) as a proxy variable to analyze its association with BMD alone. RESULTS: After adjusting for potential confounding factors, the results showed a positive correlation between lean mass and total BMD (for continuous: ß = 0.078, P < 0.001; for quartile: ß = 0.138, P < 0.001), while visceral fat mass was negatively correlated (for continuous: ß = -0.027, P < 0.001; for quartile: ß = -0.065, P < 0.001). A positive correlation was observed when the alternative variable Log LM/VFM was analyzed separately for its association with BMD (for continuous: ß = 0.034, P < 0.001; for quartile: ß = 0.084, P < 0.001). In addition, subgroup analyses for gender, age, body mass index, hypertension, and diabetes showed that all subgroups except the diabetes subgroup showed a substantial degree of robustness (P < 0.05). The smoothed curve fitting showed a nonlinear relationship between Log LM/VFM and BMD, and there was a threshold effect with a critical value of 2.60. CONCLUSION: Maintaining a proper ratio of lean body mass and visceral fat mass is beneficial for increasing BMD.

4.
Front Endocrinol (Lausanne) ; 14: 1232896, 2023.
Article in English | MEDLINE | ID: mdl-37772076

ABSTRACT

Objective: The relationship between body composition and insulin resistance (IR) is controversial. This study aimed to thoroughly examine the correlation between adipose tissue, lean body mass, and IR as evaluated by the Homeostatic Model Assessment (HOMA-IR). Methods: In this cross-sectional study, we utilized data from the National Health and Nutrition Examination Survey (NHANES) conducted between 2011 and 2018. Our study included 4981 subjects, and we employed multiple linear regression, smoothed curve fitting, threshold, and saturation effect analysis to investigate the relationship between lean body mass, visceral fat mass, and IR. Also, we used the lean body mass to visceral fat ratio (Log LM/VFM) as a proxy variable to analyze its association with IR alone. Results: The study discovered a negative link between lean body mass and IR, but the visceral fat mass was positively correlated after correcting for covariates. A negative correlation was observed when the alternative variable Log LM/VFM was analyzed separately for its association with IR. This association was present regardless of whether the exposure variables were analyzed as continuous or categorical. The data analysis revealed a nonlinear relationship between Log LM/VFM and IR, as evidenced by the generalized additive model. In addition, a threshold effect with a critical value of 1.80 and a saturation effect with a critical point of 2.5 were also observed. Further subgroup analysis for sex, age, BMI, active levels, hypertension, and diabetes showed considerable robustness between the relationship of Log LM/VFM and IR. Conclusion: Maintaining a proper ratio of lean body mass and visceral fat is beneficial for decreasing IR.

5.
Medicine (Baltimore) ; 102(10): e33194, 2023 Mar 10.
Article in English | MEDLINE | ID: mdl-36897703

ABSTRACT

BACKGROUND: Studies have shown a strong association between the triglyceride-glucose (TyG) index, a simple marker of insulin resistance, and various metabolic diseases. We performed a systematic review of the interaction between the TyG index and arterial stiffness. METHODS: Relevant observational studies assessing the association between the TyG index and arterial stiffness were thoroughly searched in PubMed, Embase, and Scopus, and a manual search of the preprint server was conducted. A random-effects model was utilized to analyze the data. The risk of bias for the included studies was assessed using the Newcastle-Ottawa Scale. A pooled effect size estimate with a random-effects model was used for the meta-analysis. RESULTS: Thirteen observational studies comprising 48,332 subjects were included. Of these, 2 were prospective cohort studies, and the remaining 11 were cross-sectional studies. According to the results of the analysis, the risk of developing high arterial stiffness was 1.85 times greater for those in the highest TyG index subgroup versus the lowest group (risk ratio [RR]: 1.85, 95% confidence interval: 1.54-2.33, I2 = 70%, P < .001). Consistent results were observed when the index was analyzed as a continuous variable (RR: 1.46, 95% confidence interval: 1.32-1.61, I2 = 77%, P < .001). A sensitivity analysis excluding each of the studies one by one yielded similar results (RRs for categorical variables: 1.67-1.94, P all <.001; RRs for continuous variables: 1.37-1.48, P all <.001). A subgroup analysis showed that different characteristics of the study subjects, such as type of study design, age, population, disease status, (including hypertension and diabetes), and pulse wave velocity measurement methods had no substantial effect on the results (P for subgroup analysis, all >0.05). CONCLUSIONS: A relatively high TyG index might be linked to an increased incidence of arterial stiffness.


Subject(s)
Insulin Resistance , Vascular Stiffness , Humans , Glucose , Triglycerides , Blood Glucose/metabolism , Risk Factors , Prospective Studies , Pulse Wave Analysis , Biomarkers
6.
BMC Plant Biol ; 22(1): 472, 2022 Oct 04.
Article in English | MEDLINE | ID: mdl-36195835

ABSTRACT

BACKGROUND: To adapt the periodic fluctuation of environmental factors, plants are subtle to monitor the natural variation for the growth and development. The daily activities and physiological functions in coordination with the natural variation are regulated by circadian clock genes. The circadian emission of floral scents is one of the rhythmic physiological activities controlled by circadian clock genes. Here, we study the molecular mechanism of circadian emission pattern of ocimene and linalool compounds in Oncidium Sharry Baby (Onc. SB) orchid. RESULTS: GC-Mass analysis revealed that Onc. SB periodically emitted ocimene and linalool during 6 to 14 o'clock daily. Terpene synthase, one of the key gene in the terpenoid biosynthetic pathway is expressed in coordination with scent emission. The promoter structure of terpene synthase revealed a circadian binding sequence (CBS), 5'-AGATTTTT-3' for CIRCADIAN CLOCK ASSOCIATED1 (CCA1) transcription factor. EMSA data confirms the binding affinity of CCA1. Transactivation assay further verified that TPS expression is regulated by CCA1. It suggests that the emission of floral scents is controlled by CCA1. CONCLUSIONS: The work validates that the mechanism of circadian emission of floral scents in Onc. Sharry Baby is controlled by the oscillator gene, CCA1(CIRCADIAN CLOCK ASSOCIATED 1) under light condition. CCA1 transcription factor up-regulates terpene synthase (TPS) by binding on CBS motif, 5'-AGATTTTT-3' of promoter region to affect the circadian emission of floral scents in Onc. SB.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Circadian Clocks , Orchidaceae , Acyclic Monoterpenes , Alkyl and Aryl Transferases , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Circadian Clocks/genetics , Circadian Rhythm/physiology , Gene Expression Regulation, Plant , Odorants , Orchidaceae/genetics , Orchidaceae/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
7.
Talanta ; 238(Pt 2): 123059, 2022 Feb 01.
Article in English | MEDLINE | ID: mdl-34808567

ABSTRACT

Hydrophilic metabolites are essential for all biological systems with multiple functions and their quantitative analysis forms an important part of metabolomics. However, poor retention of these metabolites on reversed-phase (RP) chromatographic column hinders their effective analysis with RPLC-MS methods. Herein, we developed a method for detecting hydrophilic metabolites using the ion-pair reversed-phase liquid-chromatography coupled with mass spectrometry (IPRP-LC-MS/MS) in scheduled multiple-reaction-monitoring (sMRM) mode. We first developed a hexylamine-based IPRP-UHPLC-QTOFMS method and experimentally measured retention time (tR) for 183 hydrophilic metabolites. We found that tRs of these metabolites were dominated by their electrostatic potential depending upon the numbers and types of their ionizable groups. We then systematically investigated the quantitative structure-retention relationship (QSRR) and constructed QSRR models using the measured tR. Subsequently, we developed a retention time predictive model using the random-forest regression algorithm (r2 = 0.93, q2 = 0.70, MAE = 1.28 min) for predicting metabolite retention time, which was applied in IPRP-UHPLC-MS/MS method in sMRM mode for quantitative metabolomic analysis. Our method can simultaneously quantify more than 260 metabolites. Moreover, we found that this method was applicable for multiple major biological matrices including biofluids and tissues. This approach offers an efficient method for large-scale quantitative hydrophilic metabolomic profiling even when metabolite standards are unavailable.


Subject(s)
Chromatography, Reverse-Phase , Tandem Mass Spectrometry , Chromatography, High Pressure Liquid , Chromatography, Liquid , Metabolomics
8.
Redox Biol ; 15: 266-276, 2018 05.
Article in English | MEDLINE | ID: mdl-29294438

ABSTRACT

Titanium dioxide nanoparticles (TiO2 NPs) are widely used in food and cosmetics but the health impact of human exposure remains poorly defined. Emerging evidence suggests that TiO2 NPs may elicit immune responses by acting on macrophages. Our proteomic study showed that treatment of macrophages with TiO2 NPs led to significant re-organization of cell membrane and activation of inflammation. These observations were further corroborated with transmission electron microscopy (TEM) experiments, which demonstrated that TiO2 NPs were trapped inside of multi-vesicular bodies (MVB) through endocytotic pathways. TiO2 NP caused significant mitochondrial dysfunction by increasing levels of mitochondrial reactive oxygen species (ROS), decreasing ATP generation, and decreasing metabolic flux in tricarboxylic acid (TCA) cycle from 13C-labelled glutamine using GC-MS-based metabolic flux analysis. Further lipidomic analysis showed that TiO2 NPs significantly decreased levels of cardiolipins, an important class of mitochondrial phospholipids for maintaining proper function of electron transport chains. Furthermore, TiO2 NP exposure activates inflammatory responses by increasing mRNA levels of TNF-α, iNOS, and COX-2. Consistently, our targeted metabolomic analysis showed significantly increased production of COX-2 metabolites including PGD2, PGE2, and 15d-PGJ2. In addition, TiO2 NP also caused significant attenuation of phagocytotic function of macrophages. In summary, our studies utilizing multiple powerful omic techniques suggest that human exposure of TiO2 NPs may have profound impact on macrophage function through activating inflammatory responses and causing mitochondrial dysfunction without physical presence in mitochondria.


Subject(s)
Inflammation/genetics , Mitochondria/drug effects , Nanoparticles/administration & dosage , Proteomics , Animals , Cell Membrane/drug effects , Cell Membrane/genetics , Cyclooxygenase 2/genetics , Gene Expression Regulation/drug effects , Humans , Inflammation/chemically induced , Macrophages/drug effects , Macrophages/pathology , Metabolomics , Mice , Mitochondria/pathology , Nanoparticles/chemistry , Nitric Oxide Synthase Type II/genetics , Phagocytosis/drug effects , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Titanium/administration & dosage , Titanium/chemistry , Tumor Necrosis Factor-alpha/genetics
9.
Free Radic Biol Med ; 111: 316-327, 2017 10.
Article in English | MEDLINE | ID: mdl-28456642

ABSTRACT

Mitochondrial lipids are essential for maintaining the integrity of mitochondrial membranes and the proper functions of mitochondria. As the "powerhouse" of a cell, mitochondria are also the major cellular source of reactive oxygen species (ROS). Oxidative stress occurs when the antioxidant system is overwhelmed by overproduction of ROS. Polyunsaturated fatty acids in mitochondrial membranes are primary targets for ROS attack, which may lead to lipid peroxidation (LPO) and generation of reactive lipids, such as 4-hydroxynonenal. When mitochondrial lipids are oxidized, the integrity and function of mitochondria may be compromised and this may eventually lead to mitochondrial dysfunction, which has been associated with many human diseases including cancer, cardiovascular diseases, diabetes, and neurodegenerative diseases. How mitochondrial lipids are oxidized and the underlying molecular mechanisms and pathophysiological consequences associated with mitochondrial LPO remain poorly defined. Oxidation of the mitochondria-specific phospholipid cardiolipin and generation of bioactive lipids through mitochondrial LPO has been increasingly recognized as an important event orchestrating apoptosis, metabolic reprogramming of energy production, mitophagy, and immune responses. In this review, we focus on the current understanding of how mitochondrial LPO and generation of bioactive lipid mediators in mitochondria are involved in the modulation of mitochondrial functions in the context of relevant human diseases associated with oxidative stress.


Subject(s)
Aldehydes/metabolism , Cardiovascular Diseases/metabolism , Diabetes Mellitus/metabolism , Mitochondria/metabolism , Neoplasms/metabolism , Neurodegenerative Diseases/metabolism , Animals , Apoptosis , Cardiolipins/metabolism , Cardiovascular Diseases/pathology , Diabetes Mellitus/pathology , Fatty Acids, Unsaturated/metabolism , Humans , Lipid Peroxidation , Mitochondria/pathology , Mitochondrial Membranes/metabolism , Mitochondrial Membranes/pathology , Mitophagy , Neoplasms/pathology , Neurodegenerative Diseases/pathology , Oxidative Stress , Reactive Oxygen Species/metabolism
10.
J Biol Chem ; 292(13): 5378-5391, 2017 03 31.
Article in English | MEDLINE | ID: mdl-28202546

ABSTRACT

Oxidative stress and inflammation are two major contributing factors to atherosclerosis, a leading cause of cardiovascular disease. Oxidation of phospholipids on the surface of low density lipoprotein (LDL) particles generated under oxidative stress has been associated with the progression of atherosclerosis, but the underlying molecular mechanisms remain poorly defined. We identified a novel series of oxidation products containing the cyclopentenone moiety, termed deoxy-A2/J2-isoprostanes-phosphocholine, from 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine in vivo using mass spectrometry and by comparison to a chemically synthesized standard. Transcriptomic analysis (RNA-seq) demonstrated that these compounds affected >200 genes in bone marrow-derived macrophages, and genes associated with inflammatory and anti-oxidative responses are among the top 5 differentially expressed. To further investigate the biological relevance of these novel oxidized phospholipids in atherosclerosis, we chemically synthesized a representative compound 1-palmitoyl-2-15-deoxy-δ-12,14-prostaglandin J2-sn-glycero-3-phosphocholine (15d-PGJ2-PC) and found that it induced anti-inflammatory and anti-oxidant responses in macrophages through modulation of NF-κB, peroxisome proliferator-activated receptor γ (PPARγ), and Nrf2 pathways; this compound also showed potent anti-inflammatory properties in a mice model of LPS-induced systematic inflammatory response syndrome. Additionally, 15d-PGJ2-PC inhibited macrophage foam cell formation, suggesting a beneficial role against atherosclerosis. These properties were consistent with decreased levels of these compounds in the plasma of patients with coronary heart disease compared with control subjects. Our findings uncovered a novel molecular mechanism for the negative regulation of inflammation and positive enhancement of anti-oxidative responses in macrophages by these oxidized phospholipids in LDL in the context of atherosclerosis.


Subject(s)
Anti-Inflammatory Agents/metabolism , Antioxidants/metabolism , Atherosclerosis/metabolism , Macrophages/metabolism , Phospholipids/metabolism , Animals , Anti-Inflammatory Agents/chemistry , Antioxidants/chemistry , Atherosclerosis/pathology , Cyclopentanes/metabolism , Foam Cells , Humans , Inflammation , Lipoproteins, LDL/metabolism , Mice , Mice, Transgenic , Oxidative Stress , Signal Transduction
11.
Free Radic Biol Med ; 102: 67-76, 2017 01.
Article in English | MEDLINE | ID: mdl-27838437

ABSTRACT

Altered redox status in cancer cells has been linked to lipid peroxidation induced by reactive oxygen species (ROS) and subsequent formation of reactive lipid electrophiles, especially 4-hydroxy-nonenal (4-HNE). Emerging evidence suggests that cancer cells manipulate redox status to acquire anti-apoptotic phenotype but the underlying mechanisms are poorly understood. Cardiolipin (CL), a mitochondria-specific inner membrane phospholipid, is critical for maintaining mitochondrial function. Paradoxically, liver tissues contain tetralinoleoyl cardiolipin (TLCL) as the major CL in mitochondria yet emerging evidence suggests that ROS generated in mitochondria may lead to CL peroxidation and activation of intrinsic apoptosis. It remains unclear how CL oxidation leads to apoptosis and its relevance to the pathogenesis of hepatocellular carcinoma (HCC). We employed a mass spectrometry-based lipidomic approach to profile lipids in human tissues of HCC and found that CL was gradually decreased in tumor comparing to peripheral non-cancerous tissues, accompanied by a concomitant decrease of oxidized CL and its oxidation product, 4-HNE. Incubation of liver cancer cells with TLCL significantly restored apoptotic sensitivity accompanied by an increase of CL and its oxidation products when treated with staurosporine (STS) or Sorafenib (the standard treatment for late stage HCC patients). Our studies uncovered a novel mechanism by which cancer cells adopt to evade apoptosis, highlighting the importance of mitochondrial control of apoptosis through modulation of CL oxidation and subsequent 4-HNE formation in HCC. Thus manipulation of mitochondrial CL oxidation and lipid electrophile formation may have potential therapeutic value for diseases linked to oxidative stress and mitochondrial dysfunctions.


Subject(s)
Carcinoma, Hepatocellular/genetics , Cardiolipins/metabolism , Liver Neoplasms/metabolism , Mitochondria/metabolism , Aldehydes/metabolism , Apoptosis/genetics , Carcinoma, Hepatocellular/pathology , Cardiolipins/genetics , Humans , Lipid Peroxidation/genetics , Lipids/chemistry , Lipids/genetics , Liver/metabolism , Liver/pathology , Liver Neoplasms/pathology , Mitochondria/pathology , Mitochondrial Membranes/metabolism , Mitochondrial Membranes/pathology , Oxidation-Reduction , Oxidative Stress/genetics , Reactive Oxygen Species/metabolism
12.
Front Med ; 10(2): 219-27, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27075460

ABSTRACT

A high level of social support can improve long-term diabetes self-management. Support from a single source has been evaluated. This study aims to analyze support from multiple and multilevel sources for diabetic patients by using the Chronic Illness Resources Survey (CIRS). Factors influencing the utilization of the CIRS were also evaluated. A total of 297 patients with diabetes were investigated using the CIRS and Perceived Diabetes Self-management Scale in Shihezi City, China. Descriptive statistics were used to explain demographic variables and scores of the scales. Factors affecting the utilization of chronic illness resources were determined through univariate analysis and then examined by multivariate logistic regression analysis. Of the 297 diabetic patients surveyed, 67% failed to reach the standard (more than 3 points) of utilizing chronic illness resources. Moreover, utilization of chronic illness resources was positively moderately correlated with self-management of diabetes (r = 0.75, P < 0.05). According to the multivariate logistic regression analysis, age (OR, 3.42; 95%CI, 1.19-9.84) and monthly income (OR, 5.27; 95%CI, 1.86-14.90) were significantly positively associated with the CIRS score. Individuals with high school (OR, 2.61; 95%CI, 1.13-6.05) and college (OR, 3.02; 95%CI, 1.13-8.04) degrees obtained higher scores in the survey than those with elementary school education. Results indicated that utilization of resources and support for chronic illness self-management, particularly personal adjustment and organization, were not ideal among diabetics in the communities of north-western China. Improved utilization of chronic illness resources was conducive for proper diabetes self-management. Furthermore, the level of utilization of chronic illness resources increased with age, literacy level, and monthly income.


Subject(s)
Diabetes Mellitus/therapy , Health Resources/statistics & numerical data , Self Care/standards , Social Support , Aged , Aged, 80 and over , China , Chronic Disease , Female , Humans , Logistic Models , Male , Middle Aged , Multivariate Analysis , Severity of Illness Index , Surveys and Questionnaires
13.
Redox Biol ; 4: 193-9, 2015.
Article in English | MEDLINE | ID: mdl-25598486

ABSTRACT

Oxidative stress-induced lipid peroxidation has been associated with human physiology and diseases including cancer. Overwhelming data suggest that reactive lipid mediators generated from this process, such as 4-hydroxynonenal (4-HNE), are biomarkers for oxidative stress and important players for mediating a number of signaling pathways. The biological effects of 4-HNE are primarily due to covalent modification of important biomolecules including proteins, DNA, and phospholipids containing amino group. In this review, we summarize recent progress on the role of 4-HNE in pathogenesis of cancer and focus on the involvement of mitochondria: generation of 4-HNE from oxidation of mitochondria-specific phospholipid cardiolipin; covalent modification of mitochondrial proteins, lipids, and DNA; potential therapeutic strategies for targeting mitochondrial ROS generation, lipid peroxidation, and 4-HNE.


Subject(s)
Aldehydes/metabolism , Gene Expression Regulation, Neoplastic , Lipid Peroxidation/genetics , Mitochondria/metabolism , Neoplasms/metabolism , Cardiolipins/metabolism , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Free Radicals/metabolism , Humans , Mitochondria/pathology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms/genetics , Neoplasms/pathology , Oxidative Stress , Signal Transduction
14.
Redox Biol ; 2: 878-83, 2014.
Article in English | MEDLINE | ID: mdl-25061570

ABSTRACT

Emerging evidence indicates that mitochondrial cardiolipins (CL) are prone to free radical oxidation and this process appears to be intimately associated with multiple biological functions of mitochondria. Our previous work demonstrated that a significant amount of potent lipid electrophiles including 4-hydroxy-nonenal (4-HNE) was generated from CL oxidation through a novel chemical mechanism. Here we provide further evidence that a characteristic class of CL oxidation products, epoxyalcohol-aldehyde-CL (EAA-CL), is formed through this novel mechanism in isolated mice liver mitochondria when treated with the pro-apoptotic protein t-Bid to induce cyt c release. Generation of these oxidation products are dose-dependently attenuated by a peroxidase inhibitor acetaminophen (ApAP). Using a mouse model of atherosclerosis, we detected significant amount of these CL oxidation products in liver tissue of low density lipoprotein receptor knockout (LDLR -/-) mice after Western diet feeding. Our studies highlight the importance of lipid electrophiles formation from CL oxidation in the settings of apoptosis and atherosclerosis as inhibition of CL oxidation and lipid electrophiles formation may have potential therapeutic value in diseases linked to oxidant stress and mitochondrial dysfunctions.


Subject(s)
Apoptosis , Cardiolipins/chemistry , Mitochondria/metabolism , Acetaminophen/pharmacology , Animals , Apoptosis/drug effects , Arteriosclerosis/metabolism , Arteriosclerosis/pathology , BH3 Interacting Domain Death Agonist Protein/genetics , BH3 Interacting Domain Death Agonist Protein/metabolism , BH3 Interacting Domain Death Agonist Protein/pharmacology , Cardiolipins/analysis , Chromatography, High Pressure Liquid , Diet, High-Fat , Liver/metabolism , Mass Spectrometry , Mice , Mice, Knockout , Oxidation-Reduction/drug effects , Receptors, LDL/deficiency , Receptors, LDL/genetics , Receptors, LDL/metabolism
15.
Mitochondrion ; 13(3): 209-24, 2013 May.
Article in English | MEDLINE | ID: mdl-23063711

ABSTRACT

Arachidonic acid-derived eicosanoids from cyclooxygenases, lipoxygenases, and cytochrome P450 are important lipid mediators involved in numerous homeostatic and pathophysiological processes. Most eicosanoids act primarily on their respective cell surface G-protein coupled receptors to elicit downstream signaling in an autocrine and paracrine fashion. Emerging evidence indicates that these hormones are also critical in apoptosis in a cell/tissue specific manner. In this review, we summarize the formation of eicosanoids and their roles as mediators in apoptosis, specifically on the roles of mitochondria in mediating these events and the signaling pathways involved. The biological relevance of eicosanoid-mediated apoptosis is also discussed.


Subject(s)
Apoptosis/physiology , Eicosanoids/metabolism , Mitochondria/enzymology , Oxidoreductases/metabolism , Signal Transduction/physiology , Animals , Eicosanoids/genetics , Homeostasis/physiology , Humans , Mitochondria/genetics , Oxidoreductases/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism
16.
Am J Physiol Heart Circ Physiol ; 287(2): H695-703, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15277201

ABSTRACT

Recent studies have implicated reactive oxygen species (ROS) in the pathogenesis of hypertension and activation of the sympathetic nervous system (SNS). Because nitric oxide (NO) exerts a tonic inhibition of central SNS activity, increased production of ROS could enhance inactivation of NO and result in activation of the SNS. To test the hypothesis that ROS may modulate SNS activity, we infused Tempol (4-hydroxy-2,2,6,6-tetramethyl piperidinoxyl), a superoxide dismutase mimetic, or vehicle either intravenously (250 microg x kg(-1) x min(-1)) or in the lateral ventricle (50 microg x kg body wt(-1) x min(-1)), and we determined the effects on blood pressure (BP), norepinephrine (NE) secretion from the posterior hypothalamus (PH) measured by the microdialysis technique, renal sympathetic nerve activity (RSNA) measured by direct microneurography, the abundance of neuronal NO synthase (nNOS)-mRNA in the PH, paraventricular nuclei (PVN), and locus coeruleus (LC) measured by RT-PCR, and the secretion of nitrate/nitrite (NO(x)) in the dialysate collected from the PH of Sprague-Dawley rats. Tempol reduced BP whether infused intravenously or intracerebroventricularly. Tempol reduced NE secretion from the PH and RSNA when infused intracerebroventricularly but raised NE secretion from the PH and RSNA when infused intravenously. The effects of intravenous Tempol on SNS activity were blunted or abolished by sinoaortic denervation. Tempol increased the abundance of nNOS in the PH, PVN, and LC when infused intracerebroventricularly, but it decreased the abundance of nNOS when infused intravenously. When given intracerebroventricularly, Tempol also reduced the concentration of NO(x) in the dialysate collected from the PH. Pretreatment with N(omega)-nitro-l-arginine methyl ester did not abolish the effects of intracerebral Tempol on BP, heart rate, NE secretion from the PH, and RSNA suggesting that the effects of Tempol on SNS activity may be in part dependent and in part independent of NO. In all, these studies support the notion that ROS may raise BP via activation of the SNS. This activation may be mediated in part by downregulation of nNOS and NO production, in part by mechanisms independent of NO. The discrepancy in results between intracerebroventricular and intravenous infusion of Tempol can be best explained by direct inhibitory actions on SNS activity when given intracerebral. By contrast, Tempol may exert direct vasodilation of the peripheral circulation and reflex activation of the SNS when given intravenously.


Subject(s)
Central Nervous System/physiology , Peripheral Nerves/physiology , Reactive Oxygen Species/metabolism , Sympathetic Nervous System/physiology , Animals , Blood Pressure/drug effects , Cyclic N-Oxides/pharmacology , Enzyme Inhibitors/pharmacology , Free Radical Scavengers/pharmacology , Heart Rate/drug effects , Hypothalamus, Posterior/metabolism , Interleukin-1/metabolism , Kidney/innervation , Locus Coeruleus/metabolism , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitrates/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type I , Nitrites/metabolism , Norepinephrine/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Rats , Rats, Sprague-Dawley , Spin Labels , Sympathetic Nervous System/drug effects
17.
Am J Physiol Renal Physiol ; 284(5): F1056-65, 2003 May.
Article in English | MEDLINE | ID: mdl-12554635

ABSTRACT

Renal cortical phenol injection provokes acute sympathetic nervous system-dependent hypertension and a shift of proximal tubule Na(+)/H(+) exchanger isoform 3 (NHE3) and Na(+)-P(i) cotransporter type 2 (NaPi2) to apical microvilli. This study aimed to determine whether proximal tubule (PT) Na(+) transporter redistribution persists chronically and whether the pool sizes of renal Na(+) transporters are altered. At 5 wk after a 50-microl 10% phenol injection, blood pressure is elevated: 154 +/- 8 vs. 113 +/- 11 mmHg after saline injection. Cortical membranes were fractionated into three "windows" enriched in apical brush border (WI), mixed apical and intermicrovillar cleft (WII), and intracellular membranes (WIII). NHE3 relative distribution in these windows, assessed by immunoblots and expressed as %total, remained shifted to apical from intracellular membranes (WI: 25.3 +/- 3 in phenol vs.12.7 +/- 3% in saline and WIII: 9.1 +/- 1.3 in phenol vs. 18.9 +/- 3% in saline). NaPi2 and dipeptidyl-peptidase IV also remained shifted to WI, and alkaline phosphatase activity increased 100.9 +/- 29.7 (WI) and 51.4 +/- 17.5% (WII) in phenol-injected membranes. Na(+) transporter total abundance [NHE3, NaPi2, thiazide-sensitive Na-Cl cotransporter, bumetanide-sensitive Na-K-2Cl cotransporter, Na-K-ATPase alpha(1)- and beta(1)-subunits, and epithelial Na(+) channel (ENaC) alpha- and beta-subunits] was profiled by immunoblotting. Only cortical NHE3 abundance was altered, decreasing to 0.56 +/- 0.06. The results demonstrate that phenol injury provokes a persistant shift of PT NHE3 and NaPi2 to the apical microvilli, along with a 44% decrease in total NHE3, evidence for an escape mechanism that would counteract the redistribution of a larger fraction of NHE3 to the apical surface by normalizing the total amount of NHE3 in apical membranes.


Subject(s)
Hypertension/etiology , Hypertension/metabolism , Kidney Diseases/chemically induced , Kidney Diseases/complications , Kidney/metabolism , Phenol , Sodium-Hydrogen Exchangers/metabolism , Animals , Blood Pressure/drug effects , Chronic Disease , Immunoblotting , Immunohistochemistry , Injections , Intracellular Membranes/enzymology , Kidney/drug effects , Kidney Cortex/metabolism , Kidney Medulla/metabolism , Male , Membrane Proteins/metabolism , Phenol/pharmacology , Rats , Rats, Sprague-Dawley , Sodium-Hydrogen Exchanger 3 , Sodium-Phosphate Cotransporter Proteins , Sodium-Phosphate Cotransporter Proteins, Type II , Sodium-Potassium-Exchanging ATPase/metabolism , Symporters/metabolism , Systole , Tissue Distribution
18.
Am J Hypertens ; 15(8): 717-24, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12160195

ABSTRACT

Intrarenal injection of phenol in rats causes a persistent elevation in blood pressure (BP) and in norepinephrine (NE) secretion from the posterior hypothalamus (PH), and downregulation of neuronal nitric oxide synthase (nNOS) and interleukin-1beta (IL-1beta) in the PH. These studies suggest that afferent impulses from the kidney to the brain may be responsible for hypertension associated with renal injury. Downregulation of nNOS and IL-1beta, two modulators of sympathetic nervous system (SNS) activity may mediate this activation. In this study we measured the effects of intrarenal phenol injection on peripheral SNS activity by direct renal nerve recording, plasma NE, nNOS, and IL-1beta abundance in the brain. We also determined whether renal denervation or administration of clonidine prevented these effects of phenol. Acutely, the phenol injection increased both afferent and efferent renal sympathetic nerve activity, decreased urinary sodium excretion, and increased plasma NE. Three weeks after the phenol injection, BP and plasma NE remained elevated. Renal denervation and pretreatment with clonidine prevented the increase in BP and plasma NE caused by phenol. Chronic renal injury caused by phenol was associated with decreased abundance of IL-1beta and nNOS in the PH. These studies have shown that a renal injury caused by phenol injection increases BP and central as well as peripheral SNS activity, which persist long after the injury. Renal denervation and antiadrenergic drugs abolish the effects of phenol on BP and plasma NE. Because NO and IL-1beta modulate SNS activity, the stimulatory action of phenol on the SNS could be mediated by downregulation of nNOS and IL-1beta in the brain.


Subject(s)
Afferent Pathways/drug effects , Disinfectants/adverse effects , Efferent Pathways/drug effects , Kidney Failure, Chronic/chemically induced , Kidney/drug effects , Phenol/adverse effects , Adrenergic alpha-Antagonists/therapeutic use , Animals , Clonidine/therapeutic use , Denervation , Disease Models, Animal , Disinfectants/administration & dosage , Hypertension/chemically induced , Hypothalamus, Posterior/drug effects , Injections , Interleukin-1/blood , Kidney/innervation , Kidney Failure, Chronic/prevention & control , Male , Nitric Oxide Synthase/blood , Nitric Oxide Synthase Type I , Norepinephrine/blood , Phenol/administration & dosage , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...