Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Am J Pathol ; 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38849030

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a progressive interstitial lung disease characterized by pulmonary fibroblast overactivation, resulting in the accumulation of abnormal extracellular matrix and lung parenchymal damage. Although the pathogenesis of IPF remains unclear, aging was proposed as the most prominent nongenetic risk factor. Previous studies have indicated that propionate metabolism undergoes reprogramming in the aging population, leading to the accumulation of the by-product methylmalonic acid (MMA). This study aims to explore alterations in propionate metabolism in IPF and the impact of the by-product MMA on pulmonary fibrosis. The present study revealed alterations in the expression of enzymes involved in propionate metabolism within IPF lung tissues, characterized by an increase in propionyl-CoA carboxylase and methylmalonyl-CoA epimerase expression, and a decrease in methylmalonyl-CoA mutase expression. Knockdown of methylmalonyl-CoA mutase, the key enzyme in propionate metabolism, in A549 cells induced a profibrotic phenotype and activated co-cultured fibroblasts. MMA exacerbated bleomycin-induced mouse lung fibrosis and induced a profibrotic phenotype in both epithelial cells and fibroblasts through activation of the canonical transforming growth factor-ß/Smad pathway. Overall, our findings unveil an alteration of propionate metabolism in IPF, leading to MMA accumulation, thus exacerbating lung fibrosis through promoting profibrotic phenotypic transitions via the canonical transforming growth factor-ß/Smad signaling pathway.

2.
J Transl Med ; 22(1): 479, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773615

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, fibrotic interstitial lung diseases, which mainly existed in middle-aged and elderly people. The accumulation of reactive oxygen species (ROS) is a common characteristic of IPF. Previous research also shown that lactate levels can be abnormally elevated in IPF patients. Emerging evidence suggested a relationship between lactate and ROS in IPF which needs further elucidation. In this article, we utilized a mouse model of BLM-induced pulmonary fibrosis to detect alterations in ROS levels and other indicators associated with fibrosis. Lactate could induce mitochondrial fragmentation by modulating expression and activity of DRP1 and ERK. Moreover, Increased ROS promoted P65 translocation into nucleus, leading to expression of lung fibrotic markers. Finally, Ulixertinib, Mdivi-1 and Mito-TEMPO, which were inhibitor activity of ERK, DRP1 and mtROS, respectively, could effectively prevented mitochondrial damage and production of ROS and eventually alleviate pulmonary fibrosis. Taken together, these findings suggested that lactate could promote lung fibrosis by increasing mitochondrial fission-derived ROS via ERK/DRP1 signaling, which may provide novel therapeutic solutions for IPF.


Subject(s)
Dynamins , Mice, Inbred C57BL , Mitochondrial Dynamics , Reactive Oxygen Species , Animals , Reactive Oxygen Species/metabolism , Mitochondrial Dynamics/drug effects , Dynamins/metabolism , Bleomycin , Signal Transduction , Lactic Acid/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Mitochondria/metabolism , Male , MAP Kinase Signaling System/drug effects , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Mice , Humans
3.
Int J Mol Sci ; 24(24)2023 Dec 12.
Article in English | MEDLINE | ID: mdl-38139244

ABSTRACT

The circadian rhythm is a 24 h internal clock within the body that regulates various factors, including sleep, body temperature, and hormone secretion. Circadian rhythm disruption is an important risk factor for many diseases including neurodegenerative illnesses. The central and peripheral oscillators' circadian clock network controls the circadian rhythm in mammals. The clock genes govern the central clock in the suprachiasmatic nucleus (SCN) of the brain. One function of the circadian clock is regulating lipid metabolism. However, investigations of the circadian regulation of lipid metabolism-associated apolipoprotein genes in the brain are lacking. This review summarizes the rhythmic expression of clock genes and lipid metabolism-associated apolipoprotein genes within the SCN in Mus musculus. Nine of the twenty apolipoprotein genes identified from searching the published database (SCNseq and CircaDB) are highly expressed in the SCN. Most apolipoprotein genes (ApoE, ApoC1, apoA1, ApoH, ApoM, and Cln) show rhythmic expression in the brain in mice and thus might be regulated by the master clock. Therefore, this review summarizes studies on lipid-associated apolipoprotein genes in the SCN and other brain locations, to understand how apolipoproteins associated with perturbed cerebral lipid metabolism cause multiple brain diseases and disorders. This review describes recent advancements in research, explores current questions, and identifies directions for future research.


Subject(s)
Circadian Clocks , Lipid Metabolism , Mice , Animals , Lipid Metabolism/genetics , Brain/metabolism , Circadian Rhythm/genetics , Suprachiasmatic Nucleus/metabolism , Circadian Clocks/genetics , Apolipoproteins/genetics , Apolipoproteins/metabolism , Mammals/metabolism
4.
Respir Res ; 24(1): 318, 2023 Dec 17.
Article in English | MEDLINE | ID: mdl-38105232

ABSTRACT

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a progressive interstitial lung disease (ILD) with unknown etiology, characterized by sustained damage repair of epithelial cells and abnormal activation of fibroblasts, the underlying mechanism of the disease remains elusive. METHODS: To evaluate the role of Tuftelin1 (TUFT1) in IPF and elucidate its molecular mechanism. We investigated the level of TUFT1 in the IPF and bleomycin-induced mouse models and explored the influence of TUFT1 deficiency on pulmonary fibrosis. Additionally, we explored the effect of TUFT1 on the cytoskeleton and illustrated the relationship between stress fiber and pulmonary fibrosis. RESULTS: Our results demonstrated a significant upregulation of TUFT1 in IPF and the bleomycin (BLM)-induced fibrosis model. Disruption of TUFT1 exerted inhibitory effects on pulmonary fibrosis in both in vivo and in vitro. TUFT1 facilitated the assembly of microfilaments in A549 and MRC-5 cells, with a pronounced association between TUFT1 and Neuronal Wiskott-Aldrich syndrome protein (N-WASP) observed during microfilament formation. TUFT1 can promote the phosphorylation of tyrosine residue 256 (Y256) of the N-WASP (pY256N-WASP). Furthermore, TUFT1 promoted transforming growth factor-ß1 (TGF-ß1) induced fibroblast activation by increasing nuclear translocation of pY256N-WASP in fibroblasts, while wiskostatin (Wis), an N-WASP inhibitor, suppressed these processes. CONCLUSIONS: Our findings suggested that TUFT1 plays a critical role in pulmonary fibrosis via its influence on stress fiber, and blockade of TUFT1 effectively reduces pro-fibrotic phenotypes. Pharmacological targeting of the TUFT1-N-WASP axis may represent a promising therapeutic approach for pulmonary fibrosis.


Subject(s)
Idiopathic Pulmonary Fibrosis , Lung Diseases, Interstitial , Animals , Mice , Bleomycin/toxicity , Fibroblasts/metabolism , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/metabolism , Lung/metabolism , Lung Diseases, Interstitial/metabolism , Mice, Inbred C57BL , Stress Fibers/metabolism , Transforming Growth Factor beta1/pharmacology
5.
Dalton Trans ; 52(28): 9714-9720, 2023 Jul 18.
Article in English | MEDLINE | ID: mdl-37387285

ABSTRACT

Designing an excellent OER catalyst in an alkaline environment is severe yet essential for industrial H2 application under the electrochemical technique. This study has achieved multiple modifications on CoN nanowires, the classic OER catalyst, via a facile room-temperature NaBH4 spontaneous hydrolysis. This facile process simultaneously generates oxygen vacancies and robust BN species. It wraps hydrophilic BOx motifs on the OER response CoN nanowires, producing OER active Co-N-B species, increasing active numbers and guaranteeing structural stability. It suggests that a low NaBH4 concentration (0.1 mol L-1) treatment endows CoNNWAs/CC with excellent OER performance and robust structure, which can drive a current density of 50 mA cm-2 with only 325 mV overpotentials with more than 24 hours' durability. Even, the catalyst can drive 1000 mA cm-2 around 480 mV overpotential. This study allows a novel strategy for designing high-performance OER catalysts.

6.
J Affect Disord ; 329: 72-80, 2023 05 15.
Article in English | MEDLINE | ID: mdl-36813043

ABSTRACT

BACKGROUND: Desvenlafaxine and duloxetine are selective serotonin and norepinephrine reuptake inhibitors. Their efficacy has not been directly compared using statistical hypotheses. This study evaluated the non-inferiority of desvenlafaxine extended-release (XL) to duloxetine in patients with major depressive disorder (MDD). METHODS: In this study, 420 adult patients with moderate-to-severe MDD were enrolled and randomly assigned (1:1) to receive 50 mg (once daily [QD]) of desvenlafaxine XL (n = 212) or 60 mg QD of duloxetine (n = 208). The primary endpoint was evaluated using a non-inferiority comparison based on the change from baseline to 8 weeks in the 17-item Hamilton Depression Rating Scale (HAMD17) total score. Secondary endpoints and safety were evaluated. RESULTS: Least-squares mean change in HAM-D17 total score from baseline to 8 weeks was -15.3 (95% confidence interval [CI]: -17.73, -12.89) in the desvenlafaxine XL group and - 15.9 (95% CI, -18.44, -13.39) in the duloxetine group. The least-squares mean difference was 0.6 (95% CI: -0.48, 1.69), and the upper boundary of 95% CI was less than the non-inferiority margin (2.2). No significant between-treatment differences were found in most secondary efficacy endpoints. The incidence of the most common treatment-emergent adverse events (TEAEs) was lower for desvenlafaxine XL than for duloxetine for nausea (27.2% versus 48.8%) and dizziness (18.0% versus 28.8%). LIMITATIONS: A short-term non-inferiority study without a placebo arm. CONCLUSIONS: This study demonstrated that desvenlafaxine XL 50 mg QD was non-inferior to duloxetine 60 mg QD in efficacy in patients with MDD. Desvenlafaxine had a lower incidence of TEAEs than duloxetine did.


Subject(s)
Depressive Disorder, Major , Adult , Humans , Duloxetine Hydrochloride/adverse effects , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/chemically induced , Desvenlafaxine Succinate/adverse effects , Antidepressive Agents/adverse effects , Double-Blind Method , Treatment Outcome
7.
Biomolecules ; 12(11)2022 10 24.
Article in English | MEDLINE | ID: mdl-36358897

ABSTRACT

d-Allulose is the corresponding epimer of d-fructose at the C-3 position, which exhibits a similar taste and sweetness to sucrose. As a low-calorie sweetener, d-allulose has broad application prospects in the fields of medicine, food, and so on. Currently, the production method of d-allulose is mainly the enzymatic conversion of d-fructose by d-allulose 3-epimerase (DAEase). However, the limited specific activity and thermal stability of DAEase restrict its industrial application. Herein, an ultrahigh-throughput screening assay based on the transcription factor PsiR was extensively optimized from the aspects of culture medium components, screening plasmid, and expression host, which enhanced the correction between the fluorescent readout and the enzyme activity. Then, the error-prone PCR (epPCR) library of Clostridium cellulolyticum H10 DAEase (CcDAEase) was screened through the above optimized method, and the variant I228V with improved specific activity and thermal stability was obtained. Moreover, after combining two beneficial substitutions, D281G and C289R, which were previously obtained by this optimized assay, the specific activity of the triple-mutation variant I228V/D281G/C289R reached up to 1.42-fold of the wild type (WT), while its half-life (T1/2) at 60 °C was prolonged by 62.97-fold. The results confirmed the feasibility of the optimized screening assay as a powerful tool for the directed evolution of DAEase.


Subject(s)
Fructose , Racemases and Epimerases , Racemases and Epimerases/genetics , Hydrogen-Ion Concentration , Fructose/metabolism , Protein Engineering
8.
Int J Biol Sci ; 18(15): 5681-5697, 2022.
Article in English | MEDLINE | ID: mdl-36263163

ABSTRACT

The prevalence of non-alcoholic fatty liver disease (NAFLD) increases year by year, and as a consequence, NAFLD has become one of the most prevalent liver diseases worldwide. Unfortunately, no pharmacotherapies for NAFLD have been approved by the United States Food and Drug Administration despite promising pre-clinical benefits; this situation highlights the urgent need to explore new therapeutic targets for NAFLD and for the discovery of effective therapeutic drugs. The mouse is one of the most commonly used models to study human disease and develop novel pharmacotherapies due to its small size, low-cost and ease in genetic engineering. Different mouse models are used to simulate various stages of NAFLD induced by dietary and/or genetic intervention. In this review, we summarize the newly described patho-mechanisms of NAFLD and review the preclinical mouse models of NAFLD (based on the method of induction) and appraises the use of these models in anti-NAFLD drug discovery. This article will provide a useful resource for researchers to select the appropriate model for research based on the research question being addressed.


Subject(s)
Non-alcoholic Fatty Liver Disease , United States , Mice , Humans , Animals , Non-alcoholic Fatty Liver Disease/drug therapy , Disease Models, Animal , Liver
9.
Metabolites ; 12(5)2022 May 20.
Article in English | MEDLINE | ID: mdl-35629966

ABSTRACT

The kidneys are organs that require energy from the metabolism of fatty acids and glucose; several studies have shown that the kidneys are metabolically active tissues with an estimated energy requirement similar to that of the heart. The kidneys may regulate the normal and pathological function of circulating lipids in the body, and their glomerular filtration barrier prevents large molecules or large lipoprotein particles from being filtered into pre-urine. Given the permeable nature of the kidneys, renal lipid metabolism plays an important role in affecting the rest of the body and the kidneys. Lipid metabolism in the kidneys is important because of the exchange of free fatty acids and apolipoproteins from the peripheral circulation. Apolipoproteins have important roles in the transport and metabolism of lipids within the glomeruli and renal tubules. Indeed, evidence indicates that apolipoproteins have multiple functions in regulating lipid import, transport, synthesis, storage, oxidation and export, and they are important for normal physiological function. Apolipoproteins are also risk factors for several renal diseases; for example, apolipoprotein L polymorphisms induce kidney diseases. Furthermore, renal apolipoprotein gene expression is substantially regulated under various physiological and disease conditions. This review is aimed at describing recent clinical and basic studies on the major roles and functions of apolipoproteins in the kidneys.

10.
Adv Exp Med Biol ; 1372: 119-143, 2022.
Article in English | MEDLINE | ID: mdl-35503178

ABSTRACT

High plasma levels of lipids and/or lipoproteins are risk factors for atherosclerosis, nonalcoholic fatty liver disease (NAFLD), obesity, and diabetes. These four conditions have also been identified as risk factors leading to the development of chronic kidney disease (CKD). Although many pathways that generate high plasma levels of these factors have been identified, most clinical and physiologic dysfunction results from aberrant assembly and secretion of lipoproteins. The results of several published studies suggest that elevated levels of low-density lipoprotein (LDL)-cholesterol are a risk factor for atherosclerosis, myocardial infarction, coronary artery calcification associated with type 2 diabetes, and NAFLD. Cholesterol metabolism has also been identified as an important pathway contributing to the development of CKD; clinical treatments designed to alter various steps of the cholesterol synthesis and metabolism pathway are currently under study. Cholesterol synthesis and catabolism contribute to a multistep process with pathways that are regulated at the cellular level in renal tissue. Cholesterol metabolism may also be regulated by the balance between the influx and efflux of cholesterol molecules that are capable of crossing the membrane of renal proximal tubular epithelial cells and podocytes. Cellular accumulation of cholesterol can result in lipotoxicity and ultimately kidney dysfunction and failure. Thus, further research focused on cholesterol metabolism pathways will be necessary to improve our understanding of the impact of cholesterol restriction, which is currently a primary intervention recommended for patients with dyslipidemia.


Subject(s)
Atherosclerosis , Diabetes Mellitus, Type 2 , Non-alcoholic Fatty Liver Disease , Renal Insufficiency, Chronic , Cholesterol/metabolism , Female , Humans , Lipoproteins/metabolism , Male , Renal Insufficiency, Chronic/therapy
11.
Hepatology ; 76(1): 78-93, 2022 07.
Article in English | MEDLINE | ID: mdl-34626126

ABSTRACT

BACKGROUND AND AIMS: High plasma lipid/lipoprotein levels are risk factors for various metabolic diseases. We previously showed that circadian rhythms regulate plasma lipids and deregulation of these rhythms causes hyperlipidemia and atherosclerosis in mice. Here, we show that global and liver-specific brain and muscle aryl hydrocarbon receptor nuclear translocator-like 1 (Bmal1)-deficient mice maintained on a chow or Western diet developed hyperlipidemia, denoted by the presence of higher amounts of triglyceride-rich and apolipoprotein AIV (ApoAIV)-rich larger chylomicron and VLDL due to overproduction. APPROACH AND RESULTS: Bmal1 deficiency decreased small heterodimer partner (Shp) and increased microsomal triglyceride transfer protein (MTP), a key protein that facilitates primordial lipoprotein assembly and secretion. Moreover, we show that Bmal1 regulates cAMP-responsive element-binding protein H (Crebh) to modulate ApoAIV expression and the assembly of larger lipoproteins. This is supported by the observation that Crebh-deficient and ApoAIV-deficient mice, along with Bmal1-deficient mice with knockdown of Crebh, had smaller lipoproteins. Further, overexpression of Bmal1 in Crebh-deficient mice had no effect on ApoAIV expression and lipoprotein size. CONCLUSIONS: These studies indicate that regulation of ApoAIV and assembly of larger lipoproteins by Bmal1 requires Crebh. Mechanistic studies showed that Bmal1 regulates Crebh expression by two mechanisms. First, Bmal1 interacts with the Crebh promoter to control circadian regulation. Second, Bmal1 increases Rev-erbα expression, and nuclear receptor subfamily 1 group D member 1 (Nr1D1, Rev-erbα) interacts with the Crebh promoter to repress expression. In short, Bmal1 modulates both the synthesis of primordial lipoproteins and their subsequent expansion into larger lipoproteins by regulating two different proteins, MTP and ApoAIV, through two different transcription factors, Shp and Crebh. It is likely that disruptions in circadian mechanisms contribute to hyperlipidemia and that avoiding disruptions in circadian rhythms may limit/prevent hyperlipidemia and atherosclerosis.


Subject(s)
ARNTL Transcription Factors/metabolism , Atherosclerosis , Cyclic AMP Response Element-Binding Protein/metabolism , Hyperlipidemias , Animals , Apolipoproteins A/metabolism , Atherosclerosis/metabolism , Mice , Mice, Inbred C57BL
12.
Adv Exp Med Biol ; 1276: 53-66, 2020.
Article in English | MEDLINE | ID: mdl-32705594

ABSTRACT

The basic helix-loop-helix-PAS transcription factor (CLOCK, Circadian locomotor output cycles protein kaput) was discovered in 1994 as a circadian clock. Soon after its discovery, the circadian clock, Aryl hydrocarbon receptor nuclear translocator-like protein 1 (ARNTL, also call BMAL1), was shown to regulate adiposity and body weight by controlling on the brain hypothalamic suprachiasmatic nucleus (SCN). Farther, circadian clock genes were determined to exert several of lipid metabolic and diabetes effects, overall indicating that CLOCK and BMAL1 act as a central master circadian clock. A master circadian clock acts through the neurons and hormones, with expression in the intestine, liver, kidney, lung, heart, SCN of brain, and other various cell types of the organization. Among circadian clock genes, numerous metabolic syndromes are the most important in the regulation of food intake (via regulation of circadian clock genes or clock-controlled genes in peripheral tissue), which lead to a variation in plasma phospholipids and tissue phospholipids. Circadian clock genes affect the regulation of transporters and proteins included in the regulation of phospholipid metabolism. These genes have recently received increasing recognition because a pharmacological target of circadian clock genes may be of therapeutic worth to make better resistance against insulin, diabetes, obesity, metabolism syndrome, atherosclerosis, and brain diseases. In this book chapter, we focus on the regulation of circadian clock and summarize its phospholipid effect as well as discuss the chemical, physiology, and molecular value of circadian clock pathway regulation for the treatment of plasma lipids and atherosclerosis.


Subject(s)
Circadian Clocks , Lipid Metabolism , Metabolic Diseases , ARNTL Transcription Factors , CLOCK Proteins , Circadian Clocks/genetics , Circadian Clocks/physiology , Circadian Rhythm/genetics , Circadian Rhythm/physiology , Humans , Lipid Metabolism/genetics , Metabolic Diseases/genetics , Suprachiasmatic Nucleus
13.
Mol Plant Pathol ; 21(9): 1167-1178, 2020 09.
Article in English | MEDLINE | ID: mdl-32678502

ABSTRACT

Ferritins are a large family of iron storage proteins, which are used by bacteria and other organisms to avoid iron toxicity and as a safe iron source in the cytosol. Agrobacterium tumefaciens, a phytopathogen, has two ferritin-encoding genes: atu2771 and atu2477. Atu2771 is annotated as a Bfr-encoding gene (Bacterioferritin, Bfr) and atu2477 as a Dps-encoding gene (DNA binding protein from starved cells, Dps). Three deletion mutants (Δbfr, Δdps, and bfr-dps double-deletion mutant ΔbdF) of these two ferritin-encoding genes were constructed to investigate the effects of ferritin deficiency on the iron homeostasis, oxidative stress resistance, and pathogenicity of A. tumefaciens. Deficiency of two ferritins affects the growth of A. tumefaciens under iron starvation and excess. When supplied with moderate iron, the growth of A. tumefaciens is not affected by the deficiency of ferritin. Deficiency of ferritin significantly reduces iron accumulation in the cells of A. tumefaciens, but the effect of Bfr deficiency on iron accumulation is severer than Dps deficiency and the double mutant ΔbdF has the least intracellular iron content. All three ferritin-deficient mutants showed a decreased tolerance to 3 mM H2 O2 in comparison with the wild type. The tumour induced by each of three ferritin-deficient mutants is less than that of the wild type. Complementation reversed the effects of ferritin deficiency on the growth, iron homeostasis, oxidative stress resistance, and tumorigenicity of A. tumefaciens. Therefore, ferritin plays an important role in the pathogenesis of A. tumefaciens through regulating iron homeostasis and oxidative stress survival.


Subject(s)
Agrobacterium tumefaciens/genetics , Bacterial Proteins/metabolism , Cytochrome b Group/metabolism , Ferritins/metabolism , Homeostasis , Iron/metabolism , Agrobacterium tumefaciens/pathogenicity , Agrobacterium tumefaciens/physiology , Bacterial Proteins/genetics , Cytochrome b Group/genetics , Ferritins/genetics , Hydrogen Peroxide/metabolism , Mutation , Oxidative Stress , Virulence
14.
J Clin Invest ; 130(8): 4282-4300, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32396530

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is becoming a major health issue as obesity increases around the world. We studied the effect of a circadian locomotor output cycles kaput (CLOCK) mutant (ClkΔ19/Δ19) protein on hepatic lipid metabolism in C57BL/6 Clkwt/wt and apolipoprotein E-deficient (Apoe-/-) mice. Both ClkΔ19/Δ19 and ClkΔ19/Δ19 Apoe-/- mice developed a full spectrum of liver diseases (steatosis, steatohepatitis, cirrhosis, and hepatocellular carcinoma) recognized in human NAFLD when challenged with a Western diet, lipopolysaccharide, or CoCl2. We identified induction of CD36 and hypoxia-inducible factor 1α (HIF1α) proteins as contributing factors for NAFLD. Mechanistic studies showed that WT CLOCK protein interacted with the E-box enhancer elements in the promoters of the proline hydroxylase domain (PHD) proteins to increase expression. In ClkΔ19/Δ19 mice, PHD levels were low, and HIF1α protein levels were increased. When its levels were high, HIF1α interacted with the Cd36 promoter to augment expression and enhance fatty acid uptake. Thus, these studies establish a regulatory link among circadian rhythms, hypoxia response, fatty acid uptake, and NAFLD. The mouse models described here may be useful for further mechanistic studies in the progression of liver diseases and in the discovery of drugs for the treatment of these disorders.


Subject(s)
CLOCK Proteins/metabolism , Enhancer Elements, Genetic , Mutation , Non-alcoholic Fatty Liver Disease/metabolism , Animals , CLOCK Proteins/genetics , Disease Models, Animal , Humans , Mice , Mice, Knockout, ApoE , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Protein Domains
15.
Int J Mol Sci ; 21(3)2020 Jan 28.
Article in English | MEDLINE | ID: mdl-32012883

ABSTRACT

The goal of this review was to seek a better understanding of the function and differential expression of circadian clock genes during the reproductive process. Through a discussion of how the circadian clock is involved in these steps, the identification of new clinical targets for sleep disorder-related diseases, such as reproductive failure, will be elucidated. Here, we focus on recent research findings regarding circadian clock regulation within the reproductive system, shedding new light on circadian rhythm-related problems in women. Discussions on the roles that circadian clock plays in these reproductive processes will help identify new clinical targets for such sleep disorder-related diseases.


Subject(s)
Circadian Rhythm Signaling Peptides and Proteins/metabolism , Fasting/physiology , Reproduction , Animals , Circadian Clocks , Female , Gene Expression Regulation , Humans
16.
Am J Physiol Cell Physiol ; 317(3): C492-C501, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31216190

ABSTRACT

The transcription factor aryl hydrocarbon receptor nuclear translocator-like protein-1 (BMAL1) is an essential regulator of the circadian clock, which controls the 24-h cycle of physiological processes such as nutrient absorption. To examine the role of BMAL1 in small intestinal glucose absorption, we used differentiated human colon adenocarcinoma cells (Caco-2 cells). Here, we show that BMAL1 regulates glucose uptake in differentiated Caco-2 cells and that this process is dependent on the glucose transporter sodium-glucose cotransporter 1 (SGLT1). Mechanistic studies show that BMAL1 regulates glucose uptake by controlling the transcription of SGLT1 involving paired-homeodomain transcription factor 4 (PAX4), a transcriptional repressor. This is supported by the observation that clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated endonuclease Cas9 (Cas9) knockdown of PAX4 increases SGLT1 and glucose uptake. Chromatin immunoprecipitation (ChIP) and ChIP-quantitative PCR assays show that the knockdown or overexpression of BMAL1 decreases or increases the binding of PAX4 to the hepatocyte nuclear factor 1-α binding site of the SGLT1 promoter, respectively. These findings identify BMAL1 as a critical mediator of small intestine carbohydrate absorption and SGLT1.


Subject(s)
ARNTL Transcription Factors/metabolism , Cell Differentiation/physiology , Glucose/metabolism , Homeodomain Proteins/metabolism , Paired Box Transcription Factors/metabolism , ARNTL Transcription Factors/genetics , Caco-2 Cells , Gene Knockdown Techniques , Homeodomain Proteins/genetics , Humans , Paired Box Transcription Factors/genetics
17.
Microbiol Res ; 219: 40-48, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30642465

ABSTRACT

In most signal transduction systems, coupling or scaffold proteins establish crucial connections between receptors and histidine kinases. These connections are important for signal transduction. The bacterial chemotaxis system is a canonical signal transduction system that relies on coupling proteins. The coupling proteins in the chemotaxis system have two architectures: CheW or CheV. In a typical chemotaxis signal transduction system, two CheW coupling protein molecules bridge a histidine kinase CheA dimer and two chemoreceptor (also called as methyl-accepting chemotaxis protein, MCP) trimers of dimers to form a core signaling complex and couple CheA activity to chemoreceptor control. Although CheW is a small cytoplasmic protein, it plays multiple functions in chemotaxis. CheW also builds connections between core signaling complexes, which leads to the formation of large chemosensory arrays that are responsible for collecting and amplifying signals from various chemoreceptors. Another coupling protein, CheV, shares a largely redundant ability with CheW; however, the function of CheV is not identical to that of CheW in chemotaxis. In this article, we summarize the molecular mechanism of chemotaxis in Escherichia coli and review the recent advances in the structural details and functions of CheW and CheV. Furthermore, we focus on the diversity of coupling proteins and discuss the relationship among multiple coupling proteins in one organism.


Subject(s)
Bacterial Proteins/metabolism , Chemotactic Factors/metabolism , Chemotaxis/physiology , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Histidine Kinase/metabolism , Methyl-Accepting Chemotaxis Proteins/metabolism , Amino Acid Sequence/genetics , Bacterial Proteins/genetics , Chemotactic Factors/genetics , Escherichia coli/genetics , Phosphorylation , Signal Transduction/physiology
18.
J Lipid Res ; 59(12): 2349-2359, 2018 12.
Article in English | MEDLINE | ID: mdl-30369486

ABSTRACT

Dietary fat absorption takes place in the intestine, and the liver mobilizes endogenous fat to other tissues by synthesizing lipoproteins that require apoB and microsomal triglyceride transfer protein (MTP). Dietary fat triggers the synthesis of oleoylethanolamide (OEA), a regulatory fatty acid that signals satiety to reduce food intake mainly by enhancing neural PPARα activity, in enterocytes. We explored OEA's roles in the assembly of lipoproteins in WT and Ppara-/- mouse enterocytes and hepatocytes, Caco-2 cells, and human liver-derived cells. In differentiated Caco-2 cells, OEA increased synthesis and secretion of triacylglycerols, apoB secretion in chylomicrons, and MTP expression in a dose-dependent manner. OEA also increased MTP activity and triacylglycerol secretion in WT and knockout primary enterocytes. In contrast to its intestinal cell effects, OEA reduced synthesis and secretion of triacylglycerols, apoB secretion, and MTP expression and activity in human hepatoma Huh-7 and HepG2 cells. Also, OEA reduced MTP expression and triacylglycerol secretion in WT, but not knockout, primary hepatocytes. These studies indicate differential effects of OEA on lipid synthesis and lipoprotein assembly: in enterocytes, OEA augments glycerolipid synthesis and lipoprotein assembly independent of PPARα. Conversely, in hepatocytes, OEA reduces MTP expression, glycerolipid synthesis, and lipoprotein secretion through PPARα-dependent mechanisms.


Subject(s)
Endocannabinoids/pharmacology , Intestines/drug effects , Lipoproteins/metabolism , Liver/metabolism , Oleic Acids/pharmacology , Animals , Caco-2 Cells , Carrier Proteins/metabolism , Cell Line, Tumor , Cholesterol, VLDL/metabolism , Dietary Fats/adverse effects , Enterocytes/drug effects , Enterocytes/metabolism , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Lipid Metabolism/drug effects , Liver/drug effects , Mice , PPAR alpha/metabolism
19.
J Cell Biochem ; 119(12): 9694-9706, 2018 12.
Article in English | MEDLINE | ID: mdl-30132963

ABSTRACT

OBJECTIVES: Recently, long noncoding RNAs (lncRNAs) have captured much attention for their important roles in human diseases. Deregulation of lncRNA taurine-upregulated gene 1 (TUG1) has been reported to regulate cancer progression in many cancer types. However, how TUG1 contributes to renal cell carcinoma (RCC) remains elusive; we were eager to resolve the questions. METHODS: Tumor tissues and the matched adjacent normal tissues were collected from patients with RCC. Messenger RNA (mRNA) levels of TUG1, yes-associated protein (YAP), and microRNA (miR)-9 levels were determined by reverse transcription quantitative polymerase chain reaction (RT-qPCR). The regulation of YAP by TUG1 was investigated using Western blot analysis, RT-qPCR, and immunofluorescence. The oncogenic roles of TUG1 and YAP were studied using a cell proliferation assay and a wound healing assay. The interaction of TUG1-miR-9-YAP was analyzed in RCC cell lines. RESULTS: In the current study, we observed a positive correlation between TUG1 expression and YAP expression in RCC using the Gene Expression Omnibus database and tumor tissues collected from 58 patients with RCC. The TUG1 elevation enhanced YAP expression but did not alter the Hippo-signaling pathway activity or YAP protein distribution in cells. In addition, we found that TUG1 could bind to miR-9; therefore, TUG1 could positively control YAP expression via downregulation of miR-9 level. Furthermore, we observed that inhibition of cell proliferation and cell migration induced by TUG1 silencing could be reversed by overexpression of YAP in RCC cell lines. CONCLUSIONS: Our findings indicated a pivotal role of TUG1 in driving RCC progression via regulation of miR-9/YAP, suggesting a potential therapeutic targeting role of TUG1 in RCC.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , Phosphoproteins/genetics , RNA, Long Noncoding/genetics , Adaptor Proteins, Signal Transducing/metabolism , Carcinoma, Renal Cell/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Hippo Signaling Pathway , Humans , Kidney Neoplasms/genetics , Male , MicroRNAs/genetics , Middle Aged , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Transcription Factors , YAP-Signaling Proteins
20.
Nat Commun ; 7: 13011, 2016 10 10.
Article in English | MEDLINE | ID: mdl-27721414

ABSTRACT

Circadian rhythms controlled by clock genes affect plasma lipids. Here we show that global ablation of Bmal1 in Apoe-/- and Ldlr-/- mice and its liver-specific ablation in Apoe-/- (L-Bmal1-/-Apoe-/-) mice increases, whereas overexpression of BMAL1 in L-Bmal1-/-Apoe-/- and Apoe-/-mice decreases hyperlipidaemia and atherosclerosis. Bmal1 deficiency augments hepatic lipoprotein secretion and diminishes cholesterol excretion to the bile. Further, Bmal1 deficiency reduces expression of Shp and Gata4. Reductions in Shp increase Mtp expression and lipoprotein production, whereas reductions in Gata4 diminish Abcg5/Abcg8 expression and biliary cholesterol excretion. Forced SHP expression normalizes lipoprotein secretion with no effect on biliary cholesterol excretion, while forced GATA4 expression increases cholesterol excretion to the bile and reduces plasma lipids in L-Bmal1-/-Apoe-/- and Apoe-/- mice. Thus, our data indicate that Bmal1 modulates lipoprotein production and biliary cholesterol excretion by regulating the expression of Mtp and Abcg5/Abcg8 via Shp and Gata4.


Subject(s)
ARNTL Transcription Factors/metabolism , Atherosclerosis/complications , Atherosclerosis/metabolism , Hepatocytes/metabolism , Hyperlipidemias/complications , Hyperlipidemias/metabolism , ARNTL Transcription Factors/deficiency , ATP Binding Cassette Transporter, Subfamily G, Member 5/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 5/metabolism , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/metabolism , Bile , Cell Line, Tumor , Cholesterol/metabolism , GATA4 Transcription Factor/metabolism , Humans , Lipid Metabolism , Lipoproteins/biosynthesis , Lipoproteins/genetics , Lipoproteins/metabolism , Liver/metabolism , Mice, Inbred C57BL , Models, Biological , Organ Specificity , Promoter Regions, Genetic/genetics , Protein Binding , Receptors, Cytoplasmic and Nuclear/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...