Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 53
1.
Transl Psychiatry ; 14(1): 210, 2024 May 27.
Article En | MEDLINE | ID: mdl-38802393

Atypical antipsychotics (AAPs) are primary medications for schizophrenia (SZ). However, their use is frequently associated with the development of adverse metabolic effects, and the mechanisms behind these negative effects remain inadequately elucidated. To investigate the role of macrophage migration inhibitory factor (MIF) in regulating antipsychotic-induced metabolic abnormalities, between 2017 and 2020, a cross-sectional study was conducted, involving 142 healthy individuals and 388 SZ patients undergoing treatment with either typical antipsychotic (TAP) or AAP medications. Symptoms of SZ patients were evaluated using the Positive and Negative Syndrome Scale (PANSS), and measurements of metabolic indices and plasma MIF levels were performed on all individuals. A significant increase in plasma MIF levels was observed in groups receiving five major AAP monotherapies in comparison to healthy controls (all p < 0.0001). There was no such increase shown in the group receiving TAP treatment (p > 0.05). Elevated plasma MIF levels displayed a notable correlation with insulin resistance (ß = 0.024, p = 0.020), as well as with the levels of triglycerides (ß = 0.019, p = 0.001) and total cholesterol (ß = 0.012, p = 0.038) in the groups receiving AAPs. However, while the TAP group also displayed certain metabolic dysfunction compared to healthy controls, no significant association was evident with plasma MIF levels (all p > 0.05). In conclusion, plasma MIF levels exhibit a distinctive correlation with metabolic abnormalities triggered by AAPs. Hence, there is potential for further development of MIF as a distinctive marker for monitoring adverse metabolic effects induced by AAPs in clinical settings.


Antipsychotic Agents , Intramolecular Oxidoreductases , Macrophage Migration-Inhibitory Factors , Schizophrenia , Humans , Macrophage Migration-Inhibitory Factors/blood , Male , Antipsychotic Agents/adverse effects , Antipsychotic Agents/therapeutic use , Female , Adult , Schizophrenia/drug therapy , Schizophrenia/blood , Cross-Sectional Studies , Intramolecular Oxidoreductases/blood , Middle Aged , Insulin Resistance , Case-Control Studies , Triglycerides/blood
2.
Mol Metab ; 79: 101834, 2024 Jan.
Article En | MEDLINE | ID: mdl-37935315

Attenuation of adipose hormone sensitive lipase (HSL) may impair lipolysis and exacerbate obesity. We investigate the role of cytokine, macrophage migration inhibitory factor (MIF) in regulating adipose HSL and adipocyte hypertrophy. Extracellular MIF downregulates HSL in an autocrine fashion, by activating the AMPK/JNK signaling pathway upon binding to its membrane receptor, CD74. WT mice fed high fat diet (HFD), as well as mice overexpressing MIF, both had high circulating MIF levels and showed suppression of HSL during the development of obesity. Blocking the extracellular action of MIF by a neutralizing MIF antibody significantly reduced obesity in HFD mice. Interestingly, intracellular MIF binds with COP9 signalosome subunit 5 (Csn5) and JNK, which leads to an opposing effect to inhibit JNK phosphorylation. With global MIF deletion, adipocyte JNK phosphorylation increased, resulting in decreased HSL expression, suggesting that the loss of MIF's intracellular inhibitory action on JNK was dominant in Mif-/- mice. Adipose tissue from Mif-/- mice also exhibited higher Akt and lower PKA phosphorylation following HFD feeding compared with WT, which may contribute to the downregulation of HSL activation during more severe obesity. Both intracellular and extracellular MIF have opposing effects to regulate HSL, but extracellular actions predominate to downregulate HSL and exacerbate the development of obesity during HFD.


Macrophage Migration-Inhibitory Factors , Animals , Mice , Adipocytes/metabolism , Adipose Tissue/metabolism , Macrophage Migration-Inhibitory Factors/genetics , Macrophage Migration-Inhibitory Factors/metabolism , Obesity/metabolism , Sterol Esterase/metabolism
3.
Sci Rep ; 13(1): 13068, 2023 08 11.
Article En | MEDLINE | ID: mdl-37567977

Switching to normal diet (ND) is the regular therapy for high-fat diet (HFD)-induced nonalcoholic fatty liver disease (NAFLD). Intermittent fasting (IF) is a unique treatment which may exhibits better therapeutic efficacy. Thus, we aim to investigate the therapeutic effects of these treatments and exploring the mechanisms. In the present study, NAFLD mouse model was induced by a 10-week HFD. Thereafter, mice adopted continued HFD, ND, or IF for the next 12 weeks. Finally, the liver was then harvested to assess lipid deposition, lipid metabolism, apoptosis, and autophagy, while blood was collected to determine blood glucose and insulin. The results showed that IF and ND treatment improved lipid deposition and metabolic disorder of NAFLD mice; the increasing body weight, liver weight, and HOMA-IR index of HFD mice were also alleviated by IF and ND. Furthermore, IF and ND treatment activated the macrophage migration inhibitory factor (MIF)/AMPK pathway and regulated its downstream autophagy and apoptosis. However, the efficacy of IF was better than ND. Both IF and ND activates MIF signaling and alleviate the lipotoxicity of NAFLD while IF therapy is more effective than ND. The different MIF up-regulation might be the underlying mechanism of why IF benefits more than ND.


Macrophage Migration-Inhibitory Factors , Non-alcoholic Fatty Liver Disease , Animals , Mice , Diet, High-Fat/adverse effects , Intermittent Fasting , Lipid Metabolism , Lipids , Liver/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/therapy , Non-alcoholic Fatty Liver Disease/metabolism
4.
iScience ; 26(6): 106923, 2023 Jun 16.
Article En | MEDLINE | ID: mdl-37283810

While insulin resistance (IR) is associated with inflammation in white adipose tissue, we report a non-inflammatory adipose mechanism of high fat-induced IR mediated by loss of Pref-1. Pref-1, released from adipose Pref-1+ cells with characteristics of M2 macrophages, endothelial cells or progenitors, inhibits MIF release from both Pref-1+ cells and adipocytes by binding with integrin ß1 and inhibiting the mobilization of p115. High palmitic acid induces PAR2 expression in Pref-1+ cells, downregulating Pref-1 expression and release in an AMPK-dependent manner. The loss of Pref-1 increases adipose MIF secretion contributing to non-inflammatory IR in obesity. Treatment with Pref-1 blunts the increase in circulating plasma MIF levels and subsequent IR induced by a high palmitic acid diet. Thus, high levels of fatty acids suppress Pref-1 expression and secretion, through increased activation of PAR2, resulting in an increase in MIF secretion and a non-inflammatory adipose mechanism of IR.

5.
J Control Release ; 358: 78-97, 2023 06.
Article En | MEDLINE | ID: mdl-37076016

The establishment of in vitro models of the BBB is significant for the evaluation of the mechanism and permeability of drugs and their sustained-release formulations across the BBB. Among the different models, the immortalized human cell line hCMEC/D3 has the potential to be used for a standardized in vitro BBB model due to its high throughput, reproducibility, homology and low cost. The high permeability of the paracellular pathway and the low expression of both certain transporters and metabolic enzymes in this model lead to low physiological levels of physical, transport and metabolic barriers, thus limiting the application of these cells. The barrier properties of this model have been improved in different studies by various means. However, no systematic review has been conducted on the optimization of model-building conditions or on the regulation and expression of transporters in the models. Some existing reviews focus on the overall description of the entire field of blood-brain barrier in vitro models, lacking in-depth and systematic reviews on the experimental details and model evaluation methods based on hCMEC/D3.This paper deals with a detailed review on the optimization of multiple aspects and modalities of the hCMEC/D3 cell culture process, including initial medium, optimal serum levels, Transwell membrane materials, supra-membrane supports, inoculum density, endogenous growth factor, exogenous drug levels, co-culture and transfection methods, to provide references for the establishment and evaluation of hCMEC/D3 cell models.


Blood-Brain Barrier , Endothelial Cells , Humans , Blood-Brain Barrier/metabolism , Reproducibility of Results , Endothelial Cells/metabolism , Biological Transport , Cell Line
6.
Environ Sci Pollut Res Int ; 30(7): 19304-19312, 2023 Feb.
Article En | MEDLINE | ID: mdl-36227495

Eighty 24-week-old laying hens were divided into eight groups, seven given a single oral dose per chicken with 7 dosing levels from 13.6 to 137 mg/kg body weight (bw) and one serving as sham control. The hens were observed for 28 days for clinical abnormalities, egg yield, and body weight. Egg samples from groups of low-to-medium doses were analyzed for residues of fipronil and its metabolites by LC-MS/MS. Blood and organ samples from hens of the group receiving 63.3 mg/kg bw were collected for hematochemical and histopathological analysis. We found that the median lethal dose (LD50) of fipronil was 74 mg/kg bw for laying hens. No death occurred, and there were no obvious changes in body weight and egg production in the hens receiving doses at or below 20 mg/kg bw. In the hens that survived exposure to the dose at 63.3 mg/kg bw, there was significant reduction in body weight and egg yield; histopathological changes in the liver and kidney; and increased levels of creatine, urea, glutamate oxaloacetate transferase, and glutamate pyruvic transaminase. Fipronil-sulfone was the residual marker in eggs with significantly higher concentrations and longer withdrawal periods than its maternal compound. We conclude that fipronil is efficiently transformed into fipronil-sulfone in the body with subsequent excretion into eggs. More attention should be paid to the potential food safety risk of fipronil-sulfone because of its persistence in eggs.


Chickens , Tandem Mass Spectrometry , Animals , Female , Chromatography, Liquid , Chickens/metabolism , Eggs/analysis , Body Weight , Sulfones
7.
Metabolomics ; 18(10): 76, 2022 09 30.
Article En | MEDLINE | ID: mdl-36180605

INTRODUCTION: Pro-inflammatory cytokines are responsible for initiating an effective defense against exogenous pathogens, and their regulation has a vital role in maintaining physiological homeostasis. The involvement of pro-inflammatory cytokines in pathological conditions have been explored in great detail, however, studies investigating metabolic pathways associated with these cytokines under normal homeostatic conditions are scarce. OBJECTIVES: The aim of the current study was to identify metabolites and metabolic pathways associated with circulating pro-inflammatory cytokines under homeostatic conditions using a metabolomics approach. METHODS: The study participants (n = 133) were derived from the Newfoundland Osteoarthritis Study (NFOAS) and the Complex Diseases in the Newfoundland population: Environment and Genetics (CODING) study. Plasma concentrations of cytokines including tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), interleukin-1 beta (IL-1ß), and macrophage migration inhibitory factor (MIF) were assessed by enzyme-linked immunosorbent assay. Targeted metabolomic profiling on fasting plasma samples was performed using Biocrates MxP® Quant 500 kit which measures a total of 630 metabolites. Associations between natural log-transformed metabolite concentrations and metabolite sums/ratios and cytokine levels were assessed using linear regression with adjustment for age, sex, body mass index (BMI), and osteoarthritis status. RESULTS: Seven metabolites and 11 metabolite sums/ratios were found to be significantly associated with TNF-α, IL-1ß, and MIF (all p ≤ 5.13 × 10- 5) after controlling multiple testing with Bonferroni method, indicating the association between glutathione (GSH), polyamine, and lysophosphatidylcholine (lysoPC) synthesis pathways and these pro-inflammatory cytokines. CONCLUSION: GSH, polyamine, and lysoPC synthesis pathways were positively associated with circulating TNF-α, IL-1ß, and MIF levels under homeostatic conditions.


Macrophage Migration-Inhibitory Factors , Osteoarthritis , Glutathione , Humans , Interleukin-1beta , Interleukin-6 , Lysophosphatidylcholines , Metabolomics , Polyamines , Tumor Necrosis Factor-alpha
8.
Redox Biol ; 57: 102481, 2022 Nov.
Article En | MEDLINE | ID: mdl-36148770

Metformin is widely used to surmount insulin resistance (IR) and type 2 diabetes. Accumulating evidence suggests that metformin may improve IR through regulating gut microbiota and bile acids. However, the underlying mechanisms remain unclear. Our metabolomic analysis showed that metformin significantly increased the accumulation of tauroursodeoxycholic acid (TUDCA) in intestine and liver from high-fat diet (HFD)-induced IR mice. TUDCA also alleviated IR, and reduced oxidative stress and intestinal inflammation in ob/ob mice. TUDCA blocked KEAP1 to bind with Nrf2, resulting in Nrf2 translocation into nuclear and initiating the transcription of antioxidant genes, which eventually reduced intracellular ROS accumulation and improved insulin signaling. Analysis of gut microbiota further revealed that metformin reduced the relative abundance of Bifidobacterium, which produces bile salt hydrolase (BSH). The reduction in BSH was probably crucial for the accumulation of TUDCA. Metformin also increased the proportion of Akkermanisia muciniphlia in gut microbiota of ob/ob mice via TUDCA. These beneficial effects of metformin in remodeling gut microbiota, reducing oxidative stress and improving insulin sensitivity were partly due to the accumulation of TUDCA, suggesting that TUDCA may be a potential therapy for metabolic syndrome.

9.
Metabolites ; 12(4)2022 Apr 07.
Article En | MEDLINE | ID: mdl-35448521

Obesity is a global pandemic, but there is yet no effective measure to control it. Recent metabolomics studies have identified a signature of altered amino acid profiles to be associated with obesity, but it is unclear whether these findings have actionable clinical potential. The aims of this study were to reveal the metabolic alterations of obesity and to explore potential strategies to mitigate obesity. We performed targeted metabolomic profiling of the plasma/serum samples collected from six independent cohorts and conducted an individual data meta-analysis of metabolomics for body mass index (BMI) and obesity. Based on the findings, we hypothesized that restriction of branched-chain amino acids (BCAAs), phenylalanine, or tryptophan may prevent obesity and tested our hypothesis in a dietary restriction trial with eight groups of 4-week-old male C57BL/6J mice (n = 5/group) on eight different types of diets, respectively, for 16 weeks. A total of 3397 individuals were included in the meta-analysis. The mean BMI was 30.7 ± 6.1 kg/m2, and 49% of participants were obese. Fifty-eight metabolites were associated with BMI and obesity (all p ≤ 2.58 × 10-4), linked to alterations of the BCAA, phenylalanine, tryptophan, and phospholipid metabolic pathways. The restriction of BCAAs within a high-fat diet (HFD) maintained the mice's weight, fat and lean volume, subcutaneous and visceral adipose tissue weight, and serum glucose and insulin at levels similar to those in the standard chow group, and prevented obesity, adipocyte hypertrophy, adipose inflammation, and insulin resistance induced by HFD. Our data suggest that four metabolic pathways, BCAA, phenylalanine, tryptophan, and phospholipid metabolic pathways, are altered in obesity and restriction of BCAAs within a HFD can prevent the development of obesity and insulin resistance in mice, providing a promising strategy to potentially mitigate diet-induced obesity.

10.
Curr Drug Deliv ; 19(4): 508-517, 2022.
Article En | MEDLINE | ID: mdl-34238184

BACKGROUND: The research and development of drugs for the treatment of central nervous system diseases faces many challenges at present. One of the most important questions to be answered is, how does the drug cross the blood-brain barrier to get to the target site for pharmacological action. Fluoxetine is widely used in clinical antidepressant therapy. However, the mechanism by which fluoxetine passes through the BBB also remains unclear. Under physiological pH conditions, fluoxetine is an organic cation with a relatively small molecular weight (<500), which is in line with the substrate characteristics of organic cation transporters (OCTs). Therefore, this study aimed to investigate the interaction of fluoxetine with OCTs at the BBB and BBB-associated efflux transporters. This is of great significance for fluoxetine to better treat depression. Moreover, it can provide a theoretical basis for clinical drug combination. METHODS: In vitro BBB model was developed using human brain microvascular endothelial cells (hCMEC/D3), and the cellular accumulation was tested in the presence or absence of transporter inhibitors. In addition, an in vivo trial was performed in rats to investigate the effect of OCTs on the distribution of fluoxetine in the brain tissue. Fluoxetine concentration was determined by a validated UPLC-MS/MS method. RESULTS: The results showed that amantadine (an OCT1/2 inhibitor) and prazosin (an OCT1/3 inhibitor) significantly decreased the cellular accumulation of fluoxetine (P <.001). Moreover, we found that N-methylnicotinamide (an OCT2 inhibitor) significantly inhibited the cellular uptake of 100 and 500 ng/mL fluoxetine (P <.01 and P <.05 respectively). In contrast, corticosterone (an OCT3 inhibitor) only significantly inhibited the cellular uptake of 1000 ng/mL fluoxetine (P <.05). The P-glycoprotein (P-gp) inhibitor, verapamil, and the multidrug resistance associated proteins (MRPs) inhibitor, MK571, significantly decreased the cellular uptake of fluoxetine. However, intracellular accumulation of fluoxetine was not significantly changed when fluoxetine was incubated with the breast cancer resistance protein (BCRP) inhibitor Ko143. Furthermore, in vivo experiments proved that corticosterone and prazosin significantly inhibited the brain-plasma ratio of fluoxetine at 5.5 h and 12 h, respectively. CONCLUSION: OCTs might play a significant role in the transport of fluoxetine across the BBB. In addition, P-gp, BCRP, and MRPs seemed not to mediate the efflux transport of fluoxetine.


Blood-Brain Barrier , Fluoxetine , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Animals , Biological Transport , Blood-Brain Barrier/metabolism , Chromatography, Liquid , Corticosterone/metabolism , Corticosterone/pharmacology , Endothelial Cells/metabolism , Fluoxetine/metabolism , Fluoxetine/pharmacology , Humans , Neoplasm Proteins/metabolism , Neoplasm Proteins/pharmacology , Prazosin/metabolism , Prazosin/pharmacology , Rats , Tandem Mass Spectrometry
11.
Article En | MEDLINE | ID: mdl-34358623

Inflammation is involved in the pathogenesis of psychiatric disorders. Many previous studies have defined the important roles of inflammatory factors in the pathogenesis, diagnosis, and treatment outcomes of psychiatric disorders. Macrophage migration inhibitory factor (MIF), a pro-inflammatory factor, has been gradually recognized to be involved in the development of neurological diseases in recent years. Our current review focuses on discussing the potential beneficial and detrimental roles of MIF in psychiatric disorders. We will provide new mechanistic insights for the development of potential diagnostic and therapeutic biomarkers based on MIF for psychiatric diseases.


Biomarkers , Inflammation , Macrophage Migration-Inhibitory Factors/immunology , Mental Disorders/immunology , Alzheimer Disease/immunology , Animals , Depression/immunology , Humans , Nervous System Diseases , Schizophrenia/immunology
12.
Sci Rep ; 11(1): 19116, 2021 09 27.
Article En | MEDLINE | ID: mdl-34580406

Obesity induces skeletal muscle dysfunction. The pathogenesis of which appears to substantially involve mitochondrial dysfunction, arising from impaired quality control. Exercise is a major therapeutic strategy against muscle dysfunction. Trimetazidine, a partial inhibitor of lipid oxidation, has been proposed as a metabolic modulator for several cardiovascular pathologies. However, the effects of Trimetazidine on regulating skeletal muscle function are largely unknown. Our present study used cell culture and obese mice models to test a novel hypothesis that Trimetazidine could improve muscle atrophy with similar results to exercise. In C2C12 cells, high palmitic acid-induced atrophy and mitochondrial dysfunction, which could be reversed by the treatment of Trimetazidine. In our animal models, with high-fat diet-induced obesity associated with skeletal muscle atrophy, Trimetazidine prevented muscle dysfunction, corrected metabolic abnormalities, and improved mitochondrial quality control and mitochondrial functions similarly to exercise. Thus, our study suggests that Trimetazidine successfully mimics exercise to enhance mitochondrial quality control leading to improved high-fat diet-induced muscle dysfunction.


Mitochondria/drug effects , Muscular Atrophy/therapy , Obesity/therapy , Physical Conditioning, Animal , Trimetazidine/pharmacology , Animals , Cell Line , Combined Modality Therapy , Diet, High-Fat/adverse effects , Disease Models, Animal , Humans , Lipid Metabolism/drug effects , Male , Mice , Mitochondria/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Muscular Atrophy/etiology , Muscular Atrophy/metabolism , Muscular Atrophy/physiopathology , Myoblasts , Obesity/etiology , Obesity/metabolism , Oxidation-Reduction , Palmitic Acid/toxicity , Trimetazidine/therapeutic use
13.
Front Pharmacol ; 12: 646489, 2021.
Article En | MEDLINE | ID: mdl-33935745

Muscle dysfunction is a complication of high-fat diet (HFD)-induced obesity that could be prevented by exercise, but patients did not get enough therapeutic efficacy from exercise due to multiple reasons. To explore alternative or supplementary approaches to prevent or treat muscle dysfunction in individuals with obesity, we investigated the effects of Rhodiola on muscle dysfunction as exercise pills. SIRT1 might suppress atrogenes expression and improve mitochondrial quality control, which could be a therapeutic target stimulated by exercise and Rhodiola, but further mechanisms remain unclear. We verified the lipid metabolism disorders and skeletal muscle dysfunction in HFD feeding mice. Moreover, exercise and Rhodiola were used to intervene mice with a HFD. Our results showed that exercise and Rhodiola prevented muscle atrophy and dysfunction in obese mice and activating the SIRT1 pathway, while atrogenes were suppressed and mitochondrial quality control was improved. EX-527, SIRT1 inhibitor, was used to validate the essential role of SIRT1 in salidroside benefit. Results of cell culture experiment showed that salidroside alleviated high palmitate-induced atrophy and mitochondrial quality control impairments, but these improvements of salidroside were inhibited by EX-527 in C2C12 myotubes. Overall, Rhodiola mimics exercise that activates SIRT1 signaling leading to improvement of HFD-induced muscle dysfunction.

14.
Oxid Med Cell Longev ; 2021: 6657944, 2021.
Article En | MEDLINE | ID: mdl-33791072

Long noncoding RNAs (lncRNAs) have attracted extensive attention due to their regulatory role in various cellular processes. Emerging studies have indicated that lncRNAs are expressed to varying degrees after the growth and development of the nervous system as well as injury and degeneration, thus affecting various physiological processes of the nervous system. In this review, we have compiled various reported lncRNAs related to the growth and development of central and peripheral nerves and pathophysiology (including advanced nerve centers, spinal cord, and peripheral nervous system) and explained how these lncRNAs play regulatory roles through their interactions with target-coding genes. We believe that a full understanding of the regulatory function of lncRNAs in the nervous system will contribute to understand the molecular mechanism of changes after nerve injury and will contribute to discover new diagnostic markers and therapeutic targets for nerve injury diseases.


Nervous System/metabolism , RNA, Long Noncoding/genetics , Animals , Cell Differentiation/genetics , Humans , Nerve Regeneration/genetics , Nervous System/growth & development , Nervous System Diseases/genetics , Nervous System Diseases/pathology , Neural Stem Cells/metabolism , RNA, Long Noncoding/metabolism
15.
Theranostics ; 11(10): 4710-4727, 2021.
Article En | MEDLINE | ID: mdl-33754023

Background: Telomere shortening and dysfunction may cause metabolic disorders, tissue damage and age-dependent pathologies. However, little is known about the association of telomere-associated protein Rap1 with mitochondrial energy metabolism and cardiac aging. Methods: Echocardiography was performed to detect cardiac structure and function in Rap1+/+ and Rap1-/- mice at different ages (3 months, 12 months and 20 months). Telomere length, DNA damage, cardiac senescence and cardiomyocyte size were analyzed using the real-time PCR, Western blotting, senescence associated ß-galactosidase assay and wheat germ agglutinin staining, respectively. Western blotting was also used to determine the level of cardiac fatty acid metabolism related key enzymes in mouse and human myocardium. Chromatin immunoprecipitation assay was used to verify the direct link between p53 and PPARα. The p53 inhibitor, Pifithrin-α and PPARα activator WY14643 were utilized to identify the effects of Rap1/p53/PPARα signaling pathway. Results: Telomere was shortened concomitant with extensive DNA damage in aged Rap1-/- mouse hearts, evidenced by reduced T/S ratios and increased nuclear γH2AX. Meanwhile, the aging-associated phenotypes were pronounced as reflected by altered mitochondrial ultrastructure, enhanced senescence, cardiac hypertrophy and dysfunction. Mechanistically, acetylated p53 and nuclear p53 was enhanced in the Rap1-/- mouse hearts, concomitant with reduced PPARα. Importantly, p53 directly binds to the promoter of PPARα in mouse hearts and suppresses the transcription of PPARα. In addition, aged Rap1-/- mice exhibited reduced cardiac fatty acid metabolism. Pifithrin-α alleviated cardiac aging and enhanced fatty acid metabolism in the aged Rap1-/- mice. Activating PPARα with WY14643 in primarily cultured Rap1-/- cardiomyocytes restored maximal oxygen consumption rates. Reduced Rap1 expression and impaired p53/PPARα signaling also presented in aged human myocardium. Conclusion: In summary, Rap1 may link telomere biology to fatty acid metabolism and aging-related cardiac pathologies via modulating the p53/PPARα signaling pathway, which could represent a therapeutic target in preventing/attenuating cardiac aging.


Aging/genetics , Cardiomegaly/genetics , Cellular Senescence/genetics , Myocytes, Cardiac/metabolism , PPAR alpha/genetics , Telomere-Binding Proteins/genetics , Tumor Suppressor Protein p53/genetics , Animals , Benzothiazoles/pharmacology , Cardiomegaly/diagnostic imaging , Cardiomegaly/physiopathology , DNA Damage , Echocardiography , Fatty Acids/metabolism , Heart Diseases/diagnostic imaging , Heart Diseases/genetics , Heart Diseases/physiopathology , Histones/metabolism , Mice , Mice, Knockout , Mitochondria, Heart/metabolism , Mitochondria, Heart/ultrastructure , Open Field Test , PPAR alpha/metabolism , Peroxisome Proliferators/pharmacology , Pyrimidines/pharmacology , Shelterin Complex , Signal Transduction , Telomere/metabolism , Telomere Homeostasis , Telomere-Binding Proteins/metabolism , Toluene/analogs & derivatives , Toluene/pharmacology , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism
16.
Arthritis Res Ther ; 23(1): 59, 2021 02 20.
Article En | MEDLINE | ID: mdl-33610191

BACKGROUND: Osteoarthritis (OA) is the most prevalent form of arthritis and the major cause of disability and overall diminution of quality of life in the elderly population. Currently there is no cure for OA, partly due to the large gaps in our understanding of its underlying molecular and cellular mechanisms. Macrophage migration inhibitory factor (MIF) is a procytokine that mediates pleiotropic inflammatory effects in inflammatory diseases such as rheumatoid arthritis (RA) and ankylosing spondylitis (AS). However, data on the role of MIF in OA is limited with conflicting results. We undertook this study to investigate the role of MIF in OA by examining MIF genotype, mRNA expression, and protein levels in the Newfoundland Osteoarthritis Study. METHODS: One hundred nineteen end-stage knee/hip OA patients, 16 RA patients, and 113 healthy controls were included in the study. Two polymorphisms in the MIF gene, rs755622, and -794 CATT5-8, were genotyped using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) and PCR followed by automated capillary electrophoresis, respectively. MIF mRNA levels in articular cartilage and subchondral bone were measured by quantitative polymerase chain reaction. Plasma concentrations of MIF, tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and interleukin-1 beta (IL-1ß) were measured by enzyme-linked immunosorbent assay. RESULTS: rs755622 and -794 CATT5-8 genotypes were not associated with MIF mRNA or protein levels or OA (all p ≥ 0.19). MIF mRNA level in cartilage was lower in OA patients than in controls (p = 0.028) and RA patients (p = 0.004), while the levels in bone were comparable between OA patients and controls (p = 0.165). MIF protein level in plasma was lower in OA patients than in controls (p = 3.01 × 10-10), while the levels of TNF-α, IL-6 and IL-1ß in plasma were all significantly higher in OA patients than in controls (all p ≤ 0.0007). Multivariable logistic regression showed lower MIF and higher IL-1ß protein levels in plasma were independently associated with OA (OR per SD increase = 0.10 and 8.08; 95% CI = 0.04-0.19 and 4.42-16.82, respectively), but TNF-α and IL-6 became non-significant. CONCLUSIONS: Reduced MIF mRNA and protein expression in OA patients suggested MIF might have a protective role in OA and could serve as a biomarker to differentiate OA from other joint disorders.


Arthritis, Rheumatoid , Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Osteoarthritis , Aged , Humans , Macrophage Migration-Inhibitory Factors/genetics , Quality of Life , Tumor Necrosis Factor-alpha/genetics
17.
Pain ; 162(2): 600-608, 2021 02 01.
Article En | MEDLINE | ID: mdl-32833795

ABSTRACT: Musculoskeletal pain often occurs simultaneously at multiple anatomical sites. The aim of the study was to identify metabolic biomarkers for multisite musculoskeletal pain (MSMP) by metabolomics with an extreme phenotype sampling strategy. The study participants (n = 610) were derived from the Newfoundland Osteoarthritis Study. Musculoskeletal pain was assessed using a self-reported pain questionnaire where painful sites were circled on a manikin by participants and the total number of painful sites were calculated. Targeted metabolomic profiling on fasting plasma samples was performed using the Biocrates AbsoluteIDQ p180 kit. Plasma cytokine concentrations including tumor necrosis factor-α, interleukin-6, interleukin-1ß, and macrophage migration inhibitory factor were assessed by enzyme-linked immunosorbent assay. Data on blood cholesterol profiles were retrieved from participants' medical records. Demographic, anthropological, and clinical information was self-reported. The number of reported painful sites ranged between 0 and 21. Two hundred and five participants were included in the analysis comprising 83 who had ≥7 painful sites and 122 who had ≤1 painful site. Women and younger people were more likely to have MSMP (P ≤ 0.02). Multisite musculoskeletal pain was associated with a higher risk of having incontinence, worse functional status and longer period of pain, and higher levels of low-density lipoprotein and non-high-density lipoprotein cholesterol (all P ≤ 0.03). Among the 186 metabolites measured, 2 lysophosphatidylcholines, 1 with 26 carbons with no double bond and 1 with 28 carbons with 1 double bond, were significantly and positively associated with MSMP after adjusting for multiple testing with the Bonferroni method (P ≤ 0.0001) and could be considered as novel metabolic markers for MSMP.


Musculoskeletal Pain , Female , Humans , Lysophosphatidylcholines , Metabolomics , Pain Measurement , Phenotype
18.
Oxid Med Cell Longev ; 2019: 9425183, 2019.
Article En | MEDLINE | ID: mdl-30881601

The Yiqi-Huoxue granule (YQHX) is a traditional Chinese medication widely used in the therapy of the traditional Chinese medicine diagnosis "Qi deficiency" or "blood stasis" in China. Both these symptoms are related to inflammation, but the mechanisms of YQHX against inflammation are largely unknown. Thus, our present study investigated the effects of YQHX on regulating inflammatory responses induced by lipopolysaccharides (LPS) in HUVECs. Our data found that YQHX remarkably inhibits the production of prothrombotic factors, plasminogen activator inhibitor-1 (PAI-1) and tissue factor (TF), while it upregulates the protein expression of Kruppel-like factor 2 (KLF2). The increase in PAI-1 and TF was significantly attenuated through a transgenic knockdown in KLF2 with a Lenti-shKLF2 vector. YQHX also decreases the phosphorylation of nuclear factor-κB (NF-κB) p65 and IκB following LPS stimulation, and it effectively suppresses PAI-1 and TF via a NF-κB-dependent mechanism. Taken together, our results suggest that YQHX provides a notable antithrombotic activity via regulating the KLF2 expression and NF-κB signaling pathway in HUVECs. The KLF2 and NF-κB may be potential therapeutic targets for interventions of inflammation associated with atherosclerosis.


Drugs, Chinese Herbal/therapeutic use , Kruppel-Like Transcription Factors/metabolism , NF-kappa B/metabolism , Down-Regulation , Drugs, Chinese Herbal/pharmacology , Humans , Lipopolysaccharides , Signal Transduction , Transfection
19.
J Clin Invest ; 128(11): 4997-5007, 2018 11 01.
Article En | MEDLINE | ID: mdl-30295645

Atypical antipsychotics are highly effective antischizophrenic medications but their clinical utility is limited by adverse metabolic sequelae. We investigated whether upregulation of macrophage migration inhibitory factor (MIF) underlies the insulin resistance that develops during treatment with the most commonly prescribed atypical antipsychotic, olanzapine. Olanzapine monotherapy increased BMI and circulating insulin, triglyceride, and MIF concentrations in drug-naive schizophrenic patients with normal MIF expression, but not in genotypic low MIF expressers. Olanzapine administration to mice increased their food intake and hypothalamic MIF expression, which led to activation of the appetite-related AMP-activated protein kinase and Agouti-related protein pathway. Olanzapine also upregulated MIF expression in adipose tissue, which reduced lipolysis and increased lipogenic pathways. Increased plasma lipid concentrations were associated with abnormal fat deposition in liver and skeletal muscle, which are important determinants of insulin resistance. Global MIF-gene deletion protected mice from olanzapine-induced insulin resistance, as did intracerebroventricular injection of neutralizing anti-MIF antibody, supporting the role of increased hypothalamic MIF expression in metabolic dysfunction. These findings uphold the potential pharmacogenomic value of MIF genotype determination and suggest that MIF may be a tractable target for reducing the metabolic side effects of atypical antipsychotic therapy.


Adipose Tissue/metabolism , Antipsychotic Agents/adverse effects , Hypothalamus/metabolism , Insulin Resistance , Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Olanzapine/adverse effects , Adipose Tissue/pathology , Adolescent , Adult , Animals , Antipsychotic Agents/administration & dosage , Body Mass Index , Eating/drug effects , Female , HeLa Cells , Humans , Hypothalamus/pathology , Lipids/blood , Lipolysis/drug effects , Liver/metabolism , Liver/pathology , Male , Mice , Middle Aged , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Olanzapine/administration & dosage
...