Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
PLoS Pathog ; 20(7): e1012295, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39052544

ABSTRACT

The emergence of drug-resistant Mycobacterium tuberculosis (M.tb) has led to the development of novel anti-tuberculosis (anti-TB) drugs. Common methods for testing the efficacy of new drugs, including two-dimensional cell culture models or animal models, have several limitations. Therefore, an appropriate model representative of the human organism is required. Here, we developed an M.tb infection model using human lung organoids (hLOs) and demonstrated that M.tb H37Rv can infect lung epithelial cells and human macrophages (hMφs) in hLOs. This novel M.tb infection model can be cultured long-term and split several times while maintaining a similar number of M.tb H37Rv inside the hLOs. Anti-TB drugs reduced the intracellular survival of M.tb in hLOs. Notably, M.tb growth in hLOs was effectively suppressed at each passage by rifampicin and bedaquiline. Furthermore, a reduction in inflammatory cytokine production and intracellular survival of M.tb were observed upon knockdown of MFN2 and HERPUD1 (host-directed therapeutic targets for TB) in our M.tb H37Rv-infected hLO model. Thus, the incorporation of hMφs and M.tb into hLOs provides a powerful strategy for generating an M.tb infection model. This model can effectively reflect host-pathogen interactions and be utilized to test the efficacy of anti-TB drugs and host-directed therapies.


Subject(s)
Antitubercular Agents , Lung , Mycobacterium tuberculosis , Organoids , Humans , Organoids/microbiology , Mycobacterium tuberculosis/drug effects , Lung/microbiology , Lung/pathology , Antitubercular Agents/pharmacology , Antitubercular Agents/therapeutic use , Tuberculosis, Pulmonary/drug therapy , Tuberculosis, Pulmonary/microbiology , Macrophages/microbiology , Tuberculosis/drug therapy , Tuberculosis/microbiology , Epithelial Cells/microbiology
2.
Cell Biol Toxicol ; 38(4): 557-575, 2022 08.
Article in English | MEDLINE | ID: mdl-35267148

ABSTRACT

Human lung organoids (hLOs) are useful for disease modelling and drug screening. However, a lack of immune cells in hLOs limits the recapitulation of in vivo cellular physiology. Here, we generated hLOs containing alveolar macrophage (AMφ)-like cells derived from pluripotent stem cells (PSC). To bridge hLOs with advanced human lung high-resolution X-ray computed tomography (CT), we acquired quantitative micro-CT images. Three hLO types were observed during differentiation. Among them, alveolar hLOs highly expressed not only lung epithelial cell markers but also AMφ-specific markers. Furthermore, CD68+ AMφ-like cells were spatially organized on the luminal epithelial surface of alveolar hLOs. Bleomycin-treated alveolar hLOs showed upregulated expression of fibrosis-related markers and extracellular matrix deposits in the alveolar sacs. Alveolar hLOs also showed structural alterations such as excessive tissue fraction under bleomycin treatment. Therefore, we suggest that micro-CT analyzable PSC-derived alveolar hLOs are a promising in vitro model to predict lung toxicity manifestations, including fibrosis.


Subject(s)
Pluripotent Stem Cells , Pulmonary Fibrosis , Alveolar Epithelial Cells , Bleomycin/metabolism , Humans , Lung , Macrophages, Alveolar , Organoids , Pluripotent Stem Cells/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , X-Ray Microtomography
3.
Ecotoxicol Environ Saf ; 229: 113094, 2022 Jan 01.
Article in English | MEDLINE | ID: mdl-34942421

ABSTRACT

Polyhexamethylene guanidine phosphate (PHMG-p), a humidifier disinfectant, is known to cause lung toxicity, including inflammation and pulmonary fibrosis. In this study, we aimed to investigate the effect of PHMG-p on human lung tissue models (2D epithelial cells and 3D organoids) under conditions of oxidative stress and viral infection. The effect of PHMG-p was studied by evaluating the formation of stress granules (SGs), which play a pivotal role in cellular adaptation to various stress conditions. Under oxidative stress and respiratory syncytial virus (RSV) infection, exposure to PHMG-p remarkably increased eIF2α phosphorylation, which is essential for SG-related signalling, and significantly increased SG formation. Furthermore, PHMG-p induced fibrotic gene expression and caused cell death due to severe DNA damage, which was further increased under oxidative stress and RSV infection, indicating that PHMG-p induces severe lung toxicity under stress conditions. Taken together, toxicity evaluation under various stressful conditions is necessary to accurately predict potential lung toxicity of chemicals affecting the respiratory tract.


Subject(s)
Respiratory Syncytial Virus Infections , Stress Granules , Guanidines/toxicity , Humans , Lung , Organoids
4.
Nat Chem Biol ; 12(7): 479-81, 2016 07.
Article in English | MEDLINE | ID: mdl-27159578

ABSTRACT

Increasing the thermogenic activity of adipocytes holds promise as an approach to combating human obesity and related metabolic diseases. We identified induction of mouse PR domain containing 4 (Prdm4) by the small molecule butein as a means to induce expression of uncoupling protein 1 (Ucp1), increase energy expenditure, and stimulate the generation of thermogenic adipocytes. This study highlights a Prdm4-dependent pathway, modulated by small molecules, that stimulates browning of white adipose tissue.


Subject(s)
Adipose Tissue, Brown/drug effects , Adipose Tissue, White/drug effects , Chalcones/pharmacology , DNA-Binding Proteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Animals , Chalcones/chemistry , DNA-Binding Proteins/metabolism , Diet, High-Fat/adverse effects , Mice , Mice, Inbred C57BL , Mice, Obese , Transcription Factors/metabolism
5.
Metabolism ; 157: 155938, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38795769

ABSTRACT

BACKGROUND AND AIMS: Hepatic lipogenesis is elevated in nutrient abundant conditions to convert the excess carbohydrate into triacylglycerol (TAG). Fatty acyl moiety of TAG is eventually transported into adipose tissues by very low density lipoprotein, leading to the accumulation of TAG as a preferred storage form of excess energy. Disruption of the balance between TAG clearance and synthesis leads to the accumulation of lipids in the liver, leading to the progression of non-alcoholic fatty liver disease (NAFLD) including non-alcoholic steatohepatitis. Protein arginine methyltransferase (PRMT) 6 has been linked to the various metabolic processes including hepatic gluconeogenesis, muscle atrophy and lipodystrophy in mouse models. However, the role of PRMT6 in the control of hepatic lipogenesis has not been elucidated to date. METHODS: We assessed the interaction between PRMT6 and LXR alpha by using co-immunoprecipitation assay. The specific arginine residue of LXR alpha that is methylated by PRMT6 was assessed by LC-MS/MS assay and the functional consequences of LXR alpha methylation was explored by mSREBP-1c luciferase assay. The effect of PRMT6 on hepatic lipogenesis was assessed by adenovirus-mediated ectopic expression of PRMT6 or knockdown of PRMT6 via shRNA in hepatocytes. Finally, the role of PRMT6 in hepatic lipid metabolism in vivo was explored by either ectopic expression of LXR alpha mutant that is defective in PRMT6-mediated arginine methylation or knockdown of PRMT6 in liver. RESULTS: We found that promoter activity of sterol regulatory element binding protein (SREBP) 1c is robustly activated by PRMT6. Interestingly, we demonstrated that PRMT6 binds to LXR alpha, a transcription factor for SREBP-1c, via its LXXLL motif, leading to the asymmetric dimethylation of an arginine residue and activation of this protein. Indeed, ectopic expression of PRMT6 in hepatocytes led to the enhanced expression of LXR alpha target genes in the lipogenic pathway. Conversely, genetic or pharmacological inhibition of PRMT6 diminished expression of lipogenic genes and the lipid accumulation in primary hepatocytes. Mechanistically, we found that asymmetric dimethylation of LXR alpha led to the dissociation of small heterodimer partner (SHP), a transcriptional co-inhibitor of this factor, resulting in the activation of LXR alpha-mediated transcriptional process. Finally, we showed that disruption of asymmetric dimethylation of LXR alpha in the liver led to the diminished expression of genes in the lipogenesis, resulting in the reduced hepatic lipid accumulation in high fat diet-fed mice in vivo. CONCLUSIONS: We showed that PRMT6 modulates LXR alpha activity by conferring asymmetric dimethylation of arginine 253, thus blocking SHP-mediated inhibition and promoting hepatic lipid accumulation. These results suggest that PRMT6 is critical in the control of lipid homeostasis by regulation of LXR alpha-mediated lipogenesis in the liver.


Subject(s)
Arginine , Lipogenesis , Liver X Receptors , Liver , Protein-Arginine N-Methyltransferases , Lipogenesis/genetics , Lipogenesis/physiology , Protein-Arginine N-Methyltransferases/metabolism , Protein-Arginine N-Methyltransferases/genetics , Animals , Mice , Methylation , Liver/metabolism , Arginine/metabolism , Liver X Receptors/metabolism , Liver X Receptors/genetics , Male , Humans , Hepatocytes/metabolism , Mice, Inbred C57BL , Hep G2 Cells , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 1/genetics
6.
Nat Aging ; 3(8): 982-1000, 2023 08.
Article in English | MEDLINE | ID: mdl-37488415

ABSTRACT

Adipose tissues are central in controlling metabolic homeostasis and failure in their preservation is associated with age-related metabolic disorders. The exact role of mature adipocytes in this phenomenon remains elusive. Here we describe the role of adipose branched-chain amino acid (BCAA) catabolism in this process. We found that adipocyte-specific Crtc2 knockout protected mice from age-associated metabolic decline. Multiomics analysis revealed that BCAA catabolism was impaired in aged visceral adipose tissues, leading to the activation of mechanistic target of rapamycin complex (mTORC1) signaling and the resultant cellular senescence, which was restored by Crtc2 knockout in adipocytes. Using single-cell RNA sequencing analysis, we found that age-associated decline in adipogenic potential of visceral adipose tissues was reinstated by Crtc2 knockout, via the reduction of BCAA-mTORC1 senescence-associated secretory phenotype axis. Collectively, we propose that perturbation of BCAA catabolism by CRTC2 is critical in instigating age-associated remodeling of adipose tissue and the resultant metabolic decline in vivo.


Subject(s)
Adipose Tissue , Metabolic Diseases , Mice , Animals , Adipose Tissue/metabolism , Amino Acids, Branched-Chain/metabolism , Adipocytes/metabolism , Metabolic Diseases/genetics , Mechanistic Target of Rapamycin Complex 1/genetics
7.
Environ Pollut ; 315: 120439, 2022 Dec 15.
Article in English | MEDLINE | ID: mdl-36257563

ABSTRACT

Exposure to atmospheric particulate matter (PM) increases morbidity and mortality in respiratory diseases by causing various adverse health effects; however, the effects of PM exposure on cellular stress under virus-infected conditions remain unclear. The effects of PM under 10 µm (PM10) and diesel PM (DPM) on respiratory syncytial virus (RSV) infection were investigated in human two-dimensional lung epithelial cells and human three-dimensional lung organoids mimicking the lung tissue. We evaluated the formation of stress granules, which are important in cellular adaptation to various stress conditions. Furthermore, we investigated the effects of repeated exposure to PM10 and DPM on DNA damage and cell death during viral infection. PM10 and DPM did not cause stress granule formation in the absence of RSV infection but drastically increased stress granule formation and signal transduction during RSV infection in human lung epithelial cells and human lung organoids. Further, repeated exposure to PM10 and DPM caused cell death by severely damaging DNA under RSV infection conditions. Thus, PM10 and DPM induce severe lung toxicity under stress conditions, such as viral infection, suggesting that the effects of PMs under various stressful conditions should be examined to accurately predict the lung toxicity of PM.


Subject(s)
Pneumonia, Viral , Respiratory Syncytial Virus Infections , Humans , Particulate Matter/toxicity , Organoids/metabolism , Stress Granules , Respiratory Syncytial Virus Infections/metabolism , Lung , Respiratory Syncytial Viruses
8.
Int J Syst Evol Microbiol ; 61(Pt 11): 2654-2658, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21148671

ABSTRACT

A Gram-reaction-negative, yellow-pigmented, gliding, rod-shaped, aerobic bacterium (RA5-111(T)) was isolated from foreshore soil. The taxonomic status of the novel isolate was determined using a polyphasic approach. On the basis of 16S rRNA gene sequence similarities, strain RA5-111(T) could be assigned to the genus Gramella, with sequence similarities of 97.7, 97.3 and 96.2 % to the type strains of Gramella echinicola, Gramella portivictoriae and Gramella marina, respectively. Chemotaxonomic and phenotypic characteristics also supported the affiliation of strain RA5-111(T) with the genus Gramella. The genomic DNA G+C content was 39.1 mol%. The isolate contained MK-6 as the predominant menaquinone, iso-C(15 : 0), iso-C(17 : 0) 3-OH and a summed feature (iso-C(15 : 0) 2-OH and/or C(16 : 1)ω7c) as major fatty acids, and phosphatidylethanolamine and unknown phospholipids as the polar lipids. DNA-DNA relatedness, phenotypic, genotypic and chemotaxonomic data clearly indicate that the isolate represents a novel species of the genus Gramella, for which the name Gramella gaetbulicola sp. nov. is proposed. The type strain is RA5-111(T) ( = KCTC 23022(T) = JCM 16528(T) = NBRC 106272(T)).


Subject(s)
Flavobacteriaceae/classification , Flavobacteriaceae/isolation & purification , Soil Microbiology , Base Composition , DNA, Bacterial/genetics , Fatty Acids/metabolism , Flavobacteriaceae/genetics , Flavobacteriaceae/metabolism , Molecular Sequence Data , Phylogeny , RNA, Ribosomal, 16S/genetics
9.
Anim Cells Syst (Seoul) ; 25(3): 146-151, 2021.
Article in English | MEDLINE | ID: mdl-34262657

ABSTRACT

Environmental risk factors are recognized as threats to public health. Stress granules (SGs) are non-membranous assemblies of mRNAs and proteins expressed in response to various stressors to promote cell survival. In this study, SG formation was examined to confirm the effects of polyhexamethylene guanidine phosphate (PHMG), chloromethylisothiazolinone (CMIT), and particulate matter (PM10) in airway epithelial cells, A549, HPAEpiC, and BEAS-2B cells. SGs were not observed after CMIT, PHMG, and PM10 treatments, as determined by immunofluorescence microscopy. Moreover, there was no change in the phosphorylation of the translation initiation factor eIF2αfollowing treatment with PHMG, CMIT, and PM10. Taken together, our findings might help determine the biological hazards of these materials.

10.
Diabetes ; 70(8): 1664-1678, 2021 08.
Article in English | MEDLINE | ID: mdl-34039627

ABSTRACT

Protein arginine methyltransferase (PRMT) 1 is involved in the regulation of various metabolic pathways such as glucose metabolism in liver and atrophy in the skeletal muscle. However, the role of PRMT1 in the fat tissues under the disease state has not been elucidated to date. In this study, we delineate the function of this protein in adipocytes in vivo. PRMT1 expression was abundant in the white adipose tissues (WAT), which was induced upon a high-fat diet in mice and by obesity in humans. We found that adipocyte-specific depletion of Prmt1 resulted in decreased fat mass without overall changes in body weight in mice. Mechanistically, the depletion of Prmt1 in WAT led to the activation of the AMPK pathway, which was causal to the increased lipophagy, mitochondrial lipid catabolism, and the resultant reduction in lipid droplet size in WAT in vivo. Interestingly, despite the increased energy expenditure, we observed a promotion of adipose tissue inflammation and an ectopic accumulation of triglycerides in the peripheral tissues in Prmt1 adipocyte-specific knockout mice, which promoted the impaired insulin tolerance that is reminiscent of mouse models of lipodystrophy. These data collectively suggest that PRMT1 prevents WAT from excessive degradation of triglycerides by limiting AMPK-mediated lipid catabolism to control whole-body metabolic homeostasis in diet-induced obesity conditions.


Subject(s)
Adipocytes/metabolism , Glucose/metabolism , Homeostasis/physiology , Obesity/genetics , Protein-Arginine N-Methyltransferases/genetics , 3T3-L1 Cells , Adenylate Kinase/metabolism , Adipose Tissue/metabolism , Animals , Body Weight/physiology , Diet, High-Fat , Insulin Resistance/physiology , Lipid Metabolism/physiology , Male , Mice , Mice, Knockout , Obesity/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Signal Transduction/physiology
11.
Autophagy ; 15(6): 1069-1081, 2019 06.
Article in English | MEDLINE | ID: mdl-30653406

ABSTRACT

Protein arginine methyltransferases (PRMTs) have emerged as important regulators of skeletal muscle metabolism and regeneration. However, the direct roles of the various PRMTs during skeletal muscle remodeling remain unclear. Using skeletal muscle-specific prmt1 knockout mice, we examined the function and downstream targets of PRMT1 in muscle homeostasis. We found that muscle-specific PRMT1 deficiency led to muscle atrophy. PRMT1-deficient muscles exhibited enhanced expression of a macroautophagic/autophagic marker LC3-II, FOXO3 and muscle-specific ubiquitin ligases, TRIM63/MURF-1 and FBXO32, likely contributing to muscle atrophy. The mechanistic study reveals that PRMT1 regulates FOXO3 through PRMT6 modulation. In the absence of PRMT1, increased PRMT6 specifically methylates FOXO3 at arginine 188 and 249, leading to its activation. Finally, we demonstrate that PRMT1 deficiency triggers FOXO3 hyperactivation, which is abrogated by PRMT6 depletion. Taken together, PRMT1 is a key regulator for the PRMT6-FOXO3 axis in the control of autophagy and protein degradation underlying muscle maintenance. Abbreviations: Ad-RNAi: adenovirus-delivered small interfering RNA; AKT: thymoma viral proto-oncogene; AMPK: AMP-activated protein kinase; Baf A1: bafilomycin A1; CSA: cross-sectional area; EDL: extensor digitorum longus; FBXO32: F-box protein 32; FOXO: forkhead box O; GAS: gatrocnemieus; HDAC: histone deacetylase; IGF: insulin-like growth factor; LAMP: lysosomal-associated membrane protein; MAP1LC3B/LC3B: microtubule-associated protein 1 light chain 3 beta; mKO: Mice with skeletal muscle-specific deletion of Prmt1; MTOR: mechanistic target of rapamycin kinase; MYH: myosin heavy chain; MYL1/MLC1f: myosin, light polypeptide 1; PRMT: protein arginine N-methyltransferase; sgRNA: single guide RNA; SQSTM1: sequestosome 1; SOL: soleus; TA: tibialis anterior; TRIM63/MURF-1: tripartite motif-containing 63; YY1: YY1 transcription factor.


Subject(s)
Autophagy/genetics , Forkhead Box Protein O3/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Animals , Forkhead Box Protein O3/chemistry , Forkhead Box Protein O3/genetics , HEK293 Cells , Histone Deacetylase 2/metabolism , Histone Deacetylases/metabolism , Humans , Methylation , Mice , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/pathology , Phosphorylation , Proto-Oncogene Mas , Signal Transduction/genetics , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , YY1 Transcription Factor/metabolism
12.
Nat Commun ; 9(1): 5107, 2018 11 30.
Article in English | MEDLINE | ID: mdl-30504773

ABSTRACT

Dysregulation of Ca2+/calmodulin-dependent protein kinase (CaMK)II is closely linked with myocardial hypertrophy and heart failure. However, the mechanisms that regulate CaMKII activity are incompletely understood. Here we show that protein arginine methyltransferase 1 (PRMT1) is essential for preventing cardiac CaMKII hyperactivation. Mice null for cardiac PRMT1 exhibit a rapid progression to dilated cardiomyopathy and heart failure within 2 months, accompanied by cardiomyocyte hypertrophy and fibrosis. Consistently, PRMT1 is downregulated in heart failure patients. PRMT1 depletion in isolated cardiomyocytes evokes hypertrophic responses with elevated remodeling gene expression, while PRMT1 overexpression protects against pathological responses to neurohormones. The level of active CaMKII is significantly elevated in PRMT1-deficient hearts or cardiomyocytes. PRMT1 interacts with and methylates CaMKII at arginine residues 9 and 275, leading to its inhibition. Accordingly, pharmacological inhibition of CaMKII restores contractile function in PRMT1-deficient mice. Thus, our data suggest that PRMT1 is a critical regulator of CaMKII to maintain cardiac function.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Heart Failure/etiology , Heart Failure/metabolism , Myocardium/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cell Line , Echocardiography , Electrocardiography , Electrophysiology , Heart Failure/genetics , Humans , Immunohistochemistry , Mice , Mice, Knockout , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Protein-Arginine N-Methyltransferases/genetics
13.
Mol Cells ; 45(4): 177-179, 2022 04 30.
Article in English | MEDLINE | ID: mdl-35387897
14.
Diabetes ; 65(1): 62-73, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26340929

ABSTRACT

The role of a glucagon/cAMP-dependent protein kinase-inducible coactivator PGC-1α signaling pathway is well characterized in hepatic gluconeogenesis. However, an opposing protein kinase B (PKB)/Akt-inducible corepressor signaling pathway is unknown. A previous report has demonstrated that small heterodimer partner-interacting leucine zipper protein (SMILE) regulates the nuclear receptors and transcriptional factors that control hepatic gluconeogenesis. Here, we show that hepatic SMILE expression was induced by feeding in normal mice but not in db/db and high-fat diet (HFD)-fed mice. Interestingly, SMILE expression was induced by insulin in mouse primary hepatocyte and liver. Hepatic SMILE expression was not altered by refeeding in liver-specific insulin receptor knockout (LIRKO) or PKB ß-deficient (PKBß(-/-)) mice. At the molecular level, SMILE inhibited hepatocyte nuclear factor 4-mediated transcriptional activity via direct competition with PGC-1α. Moreover, ablation of SMILE augmented gluconeogenesis and increased blood glucose levels in mice. Conversely, overexpression of SMILE reduced hepatic gluconeogenic gene expression and ameliorated hyperglycemia and glucose intolerance in db/db and HFD-fed mice. Therefore, SMILE is an insulin-inducible corepressor that suppresses hepatic gluconeogenesis. Small molecules that enhance SMILE expression would have potential for treating hyperglycemia in diabetes.


Subject(s)
Basic-Leucine Zipper Transcription Factors/genetics , Eating/genetics , Gluconeogenesis/genetics , Hepatocyte Nuclear Factor 4/genetics , Hepatocytes/metabolism , Liver/metabolism , Animals , Basic-Leucine Zipper Transcription Factors/drug effects , Basic-Leucine Zipper Transcription Factors/metabolism , Blotting, Western , Cell Line , Chromatin Immunoprecipitation , Diet, High-Fat , Gene Expression , Glucagon , Hepatocyte Nuclear Factor 4/metabolism , Hepatocytes/drug effects , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Liver/drug effects , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Polymerase Chain Reaction , Proto-Oncogene Proteins c-akt/genetics , Receptor, Insulin/genetics , Transcription Factors/genetics
15.
Endocrinol Metab (Seoul) ; 29(4): 435-40, 2014 Dec 29.
Article in English | MEDLINE | ID: mdl-25559572

ABSTRACT

Glucose homeostasis is tightly controlled by the regulation of glucose production in the liver and glucose uptake into peripheral tissues, such as skeletal muscle and adipose tissue. Under prolonged fasting, hepatic gluconeogenesis is mainly responsible for glucose production in the liver, which is essential for tissues, organs, and cells, such as skeletal muscle, the brain, and red blood cells. Hepatic gluconeogenesis is controlled in part by the concerted actions of transcriptional regulators. Fasting signals are relayed by various intracellular enzymes, such as kinases, phosphatases, acetyltransferases, and deacetylases, which affect the transcriptional activity of transcription factors and transcriptional coactivators for gluconeogenic genes. Protein arginine methyltransferases (PRMTs) were recently added to the list of enzymes that are critical for regulating transcription in hepatic gluconeogenesis. In this review, we briefly discuss general aspects of PRMTs in the control of transcription. More specifically, we summarize the roles of four PRMTs: PRMT1, PRMT 4, PRMT 5, and PRMT 6, in the control of hepatic gluconeogenesis through specific regulation of FoxO1- and CREB-dependent transcriptional events.

16.
Sci Signal ; 7(314): ra19, 2014 Feb 25.
Article in English | MEDLINE | ID: mdl-24570487

ABSTRACT

Fasting glucose homeostasis is maintained in part through cAMP (adenosine 3',5'-monophosphate)-dependent transcriptional control of hepatic gluconeogenesis by the transcription factor CREB (cAMP response element-binding protein) and its coactivator CRTC2 (CREB-regulated transcriptional coactivator 2). We showed that PRMT6 (protein arginine methyltransferase 6) promotes fasting-induced transcriptional activation of the gluconeogenic program involving CRTC2. Mass spectrometric analysis indicated that PRMT6 associated with CRTC2. In cells, PRMT6 mediated asymmetric dimethylation of multiple arginine residues of CRTC2, which enhanced the association of CRTC2 with CREB on the promoters of gluconeogenic enzyme-encoding genes. In mice, ectopic expression of PRMT6 promoted higher blood glucose concentrations, which were associated with increased expression of genes encoding gluconeogenic factors, whereas knockdown of hepatic PRMT6 decreased fasting glycemia and improved pyruvate tolerance. The abundance of hepatic PRMT6 was increased in mouse models of obesity and insulin resistance, and adenovirus-mediated depletion of PRMT6 restored euglycemia in these mice. We propose that PRMT6 is involved in the regulation of hepatic glucose metabolism in a CRTC2-dependent manner.


Subject(s)
Gluconeogenesis , Glucose/metabolism , Insulin Resistance , Liver/metabolism , Obesity/metabolism , Transcription Factors/metabolism , Animals , Arginine/genetics , Arginine/metabolism , Cell Line , Cyclic AMP/genetics , Cyclic AMP/metabolism , Glucose/genetics , Humans , Liver/pathology , Methylation , Mice , Obesity/genetics , Obesity/pathology , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Transcription Factors/genetics , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL