Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Aging Cell ; 23(2): e14049, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38062989

ABSTRACT

Subcutaneous adipose tissue (SAT), a vital energy reservoir and endocrine organ for maintaining systemic glucose, lipid, and energy homeostasis, undergoes significant changes with age. However, among the existing aging-related markers, only few genes are associated with SAT aging. In this study, weighted gene co-expression network analysis was used on a transcriptome of SAT obtained from the Genotype-Tissue Expression portal to identify biologically relevant, SAT-specific, and age-related marker genes. We found modules that exhibited significant changes with age and identified GYG2 as a novel key aging associated gene. The link between GYG2 and mitochondrial function as well as brown/beige adipocytes was supported using additional bioinformatics and experimental analyses. Additionally, we identified PPARG as the transcription factor of GYG2 expression. The newly discovered GYG2 marker can be used to not only determine the age of SAT but also uncover new mechanisms underlying SAT aging.


Subject(s)
Subcutaneous Fat , Transcriptome , Humans , Adipose Tissue/metabolism , Aging/genetics , Biomarkers/metabolism , Mitochondria/genetics , Subcutaneous Fat/metabolism , Transcriptome/genetics
2.
Front Mol Biosci ; 10: 1228640, 2023.
Article in English | MEDLINE | ID: mdl-38482540

ABSTRACT

Introduction: Senescent melanocytes are major contributors to age-related changes in the skin, highlighting the contribution to skin aging. Moreover, prolonged photodamage, such as that caused by UV exposure, can result in melanin accumulation and accelerated melanocyte senescence, thereby exacerbating aging. Melasolv™ is a substance that induces potent depigmentation effects and exhibits low toxicity. The present study aimed to investigate the potential effect of Melasolv™ on senescent melanocytes. Methods: We profiled the transcriptomics of Melasolv™-treated melanocytes and identified the possible mechanism of action (MOA) and targets using connectivity mapping analysis. We identified differentially expressed genes in response to treatment with Melasolv™ and validated the data using quantitative real-time PCR. Moreover, we performed an in vitro ß-gal assay in senescent melanocytes for further validation. Results: Melasolv™ reduced ß-gal and melanin levels in senescent melanocytes. Moreover, the identified MOAs are associated with anti-aging and anti-senescence effects. Discussion: Our findings clearly indicate that Melasolv™ not only exhibits anti-senescent properties but can also potentially alleviate melanin accumulation in senescent cells. These findings could have far-reaching implications in the treatment of age-related photodamaged skin conditions, such as senile lentigo and melasma.

3.
J Invest Dermatol ; 138(12): 2522-2530, 2018 12.
Article in English | MEDLINE | ID: mdl-29959907

ABSTRACT

Mitochondrial dysfunction can drive cellular senescence, which is accompanied by changes in metabolism and increases in senescence-associated secretory phenotypes. Although pyruvate, a key metabolite for numerous aspects of metabolism, has been used as general supplement in synthetic media, the physiological function of pyruvate underlying its protective role against cellular senescence under normal conditions has remained unknown. Here, we show that extracellular pyruvate prevents senescence in normal human dermal fibroblasts through increasing the generation of oxidized nicotinamide adenine dinucleotide (NAD+) during the conversion to lactate. Acetylated peroxisome proliferator-activated receptor gamma coactivator 1α (PGC-1α), vacuolar-type H+-ATPaseV0A1 (v-ATPaseV0A1), NF-κB p65 subunit (RelA), and histone H3 accumulate under pyruvate deprivation conditions, resulting in the onset of senescence in normal human dermal fibroblasts through the accumulation of abnormal mitochondria generated by lysosomal inactivation-induced mitophagy defects, and through an increase in senescence-associated secretory phenotypes. Furthermore, pyruvate showed a protective effect against aging phenotypes in skin equivalents, which consist of a dermis and epidermis that act similarly to in vivo skin tissues. Our findings reveal a connection between pyruvate and mitochondrial dysfunction in the progression of senescence that is, to our knowledge, previously unreported. These results suggest that the pyruvate deprivation-induced senescence model can be used to study the connection between metabolism and senescence under normal conditions.


Subject(s)
Cellular Senescence , Dermis/pathology , Epidermis/pathology , Fibroblasts/physiology , Lysosomes/metabolism , Mitochondria/metabolism , Pyruvic Acid/metabolism , Cells, Cultured , Dermis/metabolism , Epidermis/metabolism , Histones/metabolism , Humans , Ligases/metabolism , Mitochondria/pathology , Mitophagy , NAD/metabolism , PPAR gamma/metabolism
4.
Diabetes ; 65(9): 2624-38, 2016 09.
Article in English | MEDLINE | ID: mdl-27284106

ABSTRACT

Glucose-6-phosphate dehydrogenase (G6PD), a rate-limiting enzyme of the pentose phosphate pathway, plays important roles in redox regulation and de novo lipogenesis. It was recently demonstrated that aberrant upregulation of G6PD in obese adipose tissue mediates insulin resistance as a result of imbalanced energy metabolism and oxidative stress. It remains elusive, however, whether inhibition of G6PD in vivo may relieve obesity-induced insulin resistance. In this study we showed that a hematopoietic G6PD defect alleviates insulin resistance in obesity, accompanied by reduced adipose tissue inflammation. Compared with wild-type littermates, G6PD-deficient mutant (G6PD(mut)) mice were glucose tolerant upon high-fat-diet (HFD) feeding. Intriguingly, the expression of NADPH oxidase genes to produce reactive oxygen species was alleviated, whereas that of antioxidant genes was enhanced in the adipose tissue of HFD-fed G6PD(mut) mice. In diet-induced obesity (DIO), the adipose tissue of G6PD(mut) mice decreased the expression of inflammatory cytokines, accompanied by downregulated proinflammatory macrophages. Accordingly, macrophages from G6PD(mut) mice greatly suppressed lipopolysaccharide-induced proinflammatory signaling cascades, leading to enhanced insulin sensitivity in adipocytes and hepatocytes. Furthermore, adoptive transfer of G6PD(mut) bone marrow to wild-type mice attenuated adipose tissue inflammation and improved glucose tolerance in DIO. Collectively, these data suggest that inhibition of macrophage G6PD would ameliorate insulin resistance in obesity through suppression of proinflammatory responses.


Subject(s)
Adipose Tissue/metabolism , Glucosephosphate Dehydrogenase Deficiency/metabolism , Inflammation/immunology , Inflammation/metabolism , Insulin Resistance/physiology , Obesity/immunology , Obesity/metabolism , 3T3-L1 Cells , Adipocytes/metabolism , Adipose Tissue/immunology , Animals , Blotting, Western , Culture Media, Conditioned , Diet, High-Fat/adverse effects , Fasting/blood , Glucosephosphate Dehydrogenase Deficiency/genetics , Immunohistochemistry , Insulin/blood , Insulin Resistance/genetics , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Mutant Strains , Obesity/genetics , Oxidative Stress , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction
5.
Sci Rep ; 6: 28476, 2016 06 27.
Article in English | MEDLINE | ID: mdl-27346059

ABSTRACT

Lipolysis in the adipocytes provides free fatty acids for other tissues in response to the energy demand. With the rapid increase in obesity-related diseases, finding novel stimuli or mechanisms that regulate lipid metabolism becomes important. We examined the effects of visible light (410, 457, 505, 530, 590, and 660 nm) irradiation on lipolysis regulation in adipocytes differentiated from human adipose-derived stem cells (ADSCs). Interestingly, 590 nm (amber) light irradiation significantly reduced the concentration of lipid droplets (LDs). We further investigated the lipolytic signaling pathways that are involved in 590 nm light irradiation-induced breakdown of LDs. Immunoblot analysis revealed that 590 nm light irradiation-induced phosphorylation of hormone-sensitive lipase (HSL) was insufficient to promote reduction of LDs. We observed that 590 nm light irradiation decreased the expression of perilipin 1. We found that 590 nm light irradiation, but not 505 nm, induced conversion of LC3 I to LC3 II, a representative autophagic marker. We further demonstrated that the lysosomal inhibitors leupeptin/NH4Cl inhibited 590 nm light irradiation-induced reduction of LDs in differentiated adipocytes. Our data suggest that 590 nm light irradiation-induced LD breakdown is partially mediated by autophagy-related lysosomal degradation, and can be applied in clinical settings to reduce obesity.


Subject(s)
Adipocytes/physiology , Autophagy/physiology , Cell Differentiation/physiology , Lipid Droplets/physiology , Lysosomes/physiology , Adipocytes/metabolism , Amber , Cell Line , Humans , Lipase/metabolism , Lipid Droplets/metabolism , Lipid Metabolism/physiology , Lipolysis/physiology , Perilipin-1/metabolism , Phosphorylation/physiology , Signal Transduction/physiology
6.
Mol Cells ; 37(5): 365-71, 2014 May.
Article in English | MEDLINE | ID: mdl-24781408

ABSTRACT

Recent findings, notably on adipokines and adipose tissue inflammation, have revised the concept of adipose tissues being a mere storage depot for body energy. Instead, adipose tissues are emerging as endocrine and immunologically active organs with multiple effects on the regulation of systemic energy homeostasis. Notably, compared with other metabolic organs such as liver and muscle, various inflammatory responses are dynamically regulated in adipose tissues and most of the immune cells in adipose tissues are involved in obesity-mediated metabolic complications, including insulin resistance. Here, we summarize recent findings on the key roles of innate (neutrophils, macrophages, mast cells, eosinophils) and adaptive (regulatory T cells, type 1 helper T cells, CD8 T cells, B cells) immune cells in adipose tissue inflammation and metabolic dysregulation in obesity. In particular, the roles of natural killer T cells, one type of innate lymphocyte, in adipose tissue inflammation will be discussed. Finally, a new role of adipocytes as antigen presenting cells to modulate T cell activity and subsequent adipose tissue inflammation will be proposed.


Subject(s)
Adipocytes/physiology , Adipose Tissue, White/pathology , Obesity/immunology , Adipose Tissue, White/immunology , Animals , Eosinophils/physiology , Humans , Inflammation/metabolism , Insulin Resistance/immunology , Lymphocytes/physiology , Macrophages/physiology , Mast Cells/physiology , Neutrophils/physiology , Obesity/pathology
7.
Mol Cell Biol ; 33(12): 2425-35, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23572562

ABSTRACT

Glucose-6-phosphate dehydrogenase (G6PD) is a key enzyme that regulates cellular redox potential. In this study, we demonstrate that macrophage G6PD plays an important role in the modulation of proinflammatory responses and oxidative stress. The G6PD levels in macrophages in the adipose tissue of obese animals were elevated, and G6PD mRNA levels positively correlated with those of proinflammatory genes. Lipopolysaccharide (LPS) and free fatty acids, which initiate proinflammatory signals, stimulated macrophage G6PD. Overexpression of macrophage G6PD potentiated the expression of proinflammatory and pro-oxidative genes responsible for the aggravation of insulin sensitivity in adipocytes. In contrast, when macrophage G6PD was inhibited or suppressed via chemical inhibitors or small interfering RNA (siRNA), respectively, basal and LPS-induced proinflammatory gene expression was attenuated. Furthermore, macrophage G6PD increased activation of the p38 mitogen-activated protein kinase (MAPK) and NF-κB pathways, which may lead to a vicious cycle of oxidative stress and proinflammatory cascade. Together, these data suggest that an abnormal increase of G6PD in macrophages promotes oxidative stress and inflammatory responses in the adipose tissue of obese animals.


Subject(s)
Glucosephosphate Dehydrogenase/metabolism , Macrophages/metabolism , Oxidative Stress , Adipocytes/metabolism , Adipose Tissue/enzymology , Adipose Tissue/metabolism , Animals , Cell Line , Chemokine CCL2/biosynthesis , Fatty Acids, Nonesterified/metabolism , Female , Glucosephosphate Dehydrogenase/antagonists & inhibitors , Glucosephosphate Dehydrogenase/genetics , Green Fluorescent Proteins/genetics , Humans , Inflammation/immunology , Interleukin-1beta/biosynthesis , Interleukin-6/biosynthesis , Lipopolysaccharides/pharmacology , Macrophages/enzymology , Male , Mice , Mice, Inbred C57BL , NADP/pharmacology , NF-kappa B/metabolism , Obesity , Oxidation-Reduction , RNA Interference , RNA, Messenger/analysis , RNA, Small Interfering , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Diabetes ; 60(10): 2474-83, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21911747

ABSTRACT

OBJECTIVE: Tissue inflammation is a key factor underlying insulin resistance in established obesity. Several models of immuno-compromised mice are protected from obesity-induced insulin resistance. However, it is unanswered whether inflammation triggers systemic insulin resistance or vice versa in obesity. The purpose of this study was to assess these questions. RESEARCH DESIGN AND METHODS: We fed a high-fat diet (HFD) to wild-type mice and three different immuno-compromised mouse models (lymphocyte-deficient Rag1 knockout, macrophage-depleted, and hematopoietic cell-specific Jun NH(2)-terminal kinase-deficient mice) and measured the time course of changes in macrophage content, inflammatory markers, and lipid accumulation in adipose tissue, liver, and skeletal muscle along with systemic insulin sensitivity. RESULTS: In wild-type mice, body weight and adipose tissue mass, as well as insulin resistance, were clearly increased by 3 days of HFD. Concurrently, in the short-term HFD period inflammation was selectively elevated in adipose tissue. Interestingly, however, all three immuno-compromised mouse models were not protected from insulin resistance induced by the short-term HFD. On the other hand, lipid content was markedly increased in liver and skeletal muscle at day 3 of HFD. CONCLUSIONS: These data suggest that the initial stage of HFD-induced insulin resistance is independent of inflammation, whereas the more chronic state of insulin resistance in established obesity is largely mediated by macrophage-induced proinflammatory actions. The early-onset insulin resistance during HFD feeding is more likely related to acute tissue lipid overload.


Subject(s)
Dietary Fats/administration & dosage , Dietary Fats/adverse effects , Inflammation/chemically induced , Inflammation/metabolism , Insulin Resistance/physiology , Adipose Tissue/metabolism , Animals , Blood Glucose , Ceramides/metabolism , Drug Administration Schedule , Epididymis/metabolism , Glucose/metabolism , Glucose Tolerance Test , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism
9.
Endocrinology ; 152(3): 793-803, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21248143

ABSTRACT

Increased reactive oxygen species (ROS) induce pancreatic ß-cell dysfunction during progressive type 2 diabetes. Glucose-6-phosphate dehydrogenase (G6PD) is a reduced nicotinamide adenine dinucleotide phosphate-producing enzyme that plays a key role in cellular reduction/oxidation regulation. We have investigated whether variations in G6PD contribute to ß-cell dysfunction through regulation of ROS accumulation and ß-cell gene expression. When the level of G6PD expression in pancreatic islets was examined in several diabetic animal models, such as db/db mice and OLEFT rats, G6PD expression was evidently up-regulated in pancreatic islets in diabetic animals. To investigate the effect of G6PD on ß-cell dysfunction, we assessed the levels of cellular ROS, glucose-stimulated insulin secretion and ß-cell apoptosis in G6PD-overexpressing pancreatic ß-cells. In INS-1 cells, G6PD overexpression augmented ROS accumulation associated with increased expression of prooxidative enzymes, such as inducible nitric oxide synthase and reduced nicotinamide adenine dinucleotide phosphate oxidase. G6PD up-regulation also caused decrease in glucose-stimulated insulin secretion in INS-1 cells and primary pancreatic islets. Moreover, elevated G6PD expression led to ß-cell apoptosis, concomitant with the increase in proapoptotic gene expression. On the contrary, suppression of G6PD with small interference RNA attenuated palmitate-induced ß-cell apoptosis. Together, these data suggest that up-regulation of G6PD in pancreatic ß-cells would induce ß-cell dysregulation through ROS accumulation in the development of type 2 diabetes.


Subject(s)
Gene Expression Regulation, Enzymologic/physiology , Glucosephosphate Dehydrogenase/metabolism , Insulin-Secreting Cells/metabolism , Up-Regulation , Animals , Apoptosis , Cell Line, Tumor , Glucose , Glucosephosphate Dehydrogenase/genetics , Insulin/metabolism , Insulin-Secreting Cells/cytology , Mice , Mice, Inbred NOD , Palmitates , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species
10.
J Lipid Res ; 51(12): 3425-33, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20847297

ABSTRACT

Liver X receptor (LXR), a sterol-activated nuclear hormone receptor, has been implicated in cholesterol and fatty acid homeostasis via regulation of reverse cholesterol transport and de novo fatty acid synthesis. LXR is also involved in immune responses, including anti-inflammatory action and T cell proliferation. In this study, we demonstrated that activated LXR suppresses cell cycle progression and proliferation in certain cell types. Stimulation of LXR with synthetic ligand T0901317 or GW3965 inhibited cell growth rate and arrested the cell cycle at the G1/S boundary in several cells, such as RWPE1, THP1, SNU16, LNCaP, and HepG2. However, LXR ligands did not exhibit antiproliferative activity in PC3, HEK293, or HeLa cells. Interestingly, activated LXR-mediated cell cycle arrest is closely correlated with the lipogenic gene expression and triacylglyceride accumulation. In accordance with these findings, suppression of FAS via small-interference RNA (siRNA) partially alleviated the antiproliferative effect of LXR activation in RWPE1 cells. Together, these data suggest that LXR activation with its ligands inhibits cell proliferation and induces G1/S arrest through elevated lipogenic activity, thus proposing a novel effect of activated LXR on cell cycle regulation.


Subject(s)
Benzoates/pharmacology , Benzylamines/pharmacology , Cell Cycle/drug effects , Cell Proliferation/drug effects , Hydrocarbons, Fluorinated/pharmacology , Lipogenesis/drug effects , Orphan Nuclear Receptors/agonists , Orphan Nuclear Receptors/metabolism , Sulfonamides/pharmacology , Dose-Response Relationship, Drug , Fatty Acids/biosynthesis , Fatty Acids/metabolism , Humans , Ligands , Liver X Receptors , Tumor Cells, Cultured
11.
Metabolism ; 59(9): 1241-51, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20045538

ABSTRACT

Adipose tissue expresses all components of the renin-angiotensin system including angiotensinogen (AGT). Recent studies have highlighted a potential role of AGT in adipose tissue function and homeostasis. However, some controversies surround the regulatory mechanisms of AGT in obese adipose tissue. In this context, we here demonstrated that the AGT messenger RNA (mRNA) level in human subcutaneous adipose tissue was significantly reduced in obese subjects as compared with nonobese subjects. Adipose tissue AGT mRNA level in obese mice was also lower as compared with their lean littermates; however, the hepatic AGT mRNA level remained unchanged. When 3T3-L1 adipocytes were cultured for a long period, the adipocytes became hypertrophic with a marked increase in the production of reactive oxygen species. Expression and secretion of AGT continued to decrease during the course of adipocyte hypertrophy. Treatment of the 3T3-L1 and primary adipocytes with reactive oxygen species (hydrogen peroxide) or tumor necrosis factor alpha caused a significant decrease in the expression and secretion of AGT. On the other hand, treatment with the antioxidant N-acetyl cysteine suppressed the decrease in the expression and secretion of AGT in the hypertrophied 3T3-L1 adipocytes. Finally, treatment of obese db/db mice with N-acetyl cysteine augmented the expression of AGT in the adipose tissue, but not in the liver. The present study demonstrates for the first time that oxidative stress dysregulates AGT in obese adipose tissue, providing a novel insight into the adipose tissue-specific interaction between the regulation of AGT and oxidative stress in the pathophysiology of obesity.


Subject(s)
Adipocytes/metabolism , Angiotensinogen/metabolism , Obesity/metabolism , Subcutaneous Fat/metabolism , Adipocytes/cytology , Adult , Angiotensinogen/genetics , Animals , Cell Size , Cells, Cultured , Female , Humans , Male , Mice , Mice, Obese , Middle Aged , Obesity/genetics , Obesity/physiopathology , Oxidative Stress , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Subcutaneous Fat/physiopathology , Tumor Necrosis Factor-alpha/metabolism
12.
Am J Physiol Endocrinol Metab ; 296(4): E955-64, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19208854

ABSTRACT

Berberine (BBR) has been shown to improve several metabolic disorders, such as obesity, type 2 diabetes, and dyslipidemia, by stimulating AMP-activated protein kinase (AMPK). However, the effects of BBR on proinflammatory responses in macrophages are poorly understood. Here we show that BBR represses proinflammatory responses through AMPK activation in macrophages. In adipose tissue of obese db/db mice, BBR treatment significantly downregulated the expression of proinflammatory genes such as TNF-alpha, IL-1beta, IL-6, monocyte chemoattractant protein-1 (MCP-1), inducible nitric oxide synthase (iNOS), and cyclooxygenase-2 (COX-2). Consistently, BBR inhibited LPS-induced expression of proinflammatory genes including IL-1beta, IL-6, iNOS, MCP-1, COX-2, and matrix metalloprotease-9 in peritoneal macrophages and RAW 264.7 cells. Upon various proinflammatory signals including LPS, free fatty acids, and hydrogen peroxide, BBR suppressed the phosphorylation of MAPKs, such as p38, ERK, and JNK, and the level of reactive oxygen species in macrophages. Moreover, these inhibitory effects of BBR on proinflammatory responses were abolished by AMPK inhibition via either compound C, an AMPK inhibitor, or dominant-negative AMPK, implying that BBR would downregulate proinflammatory responses in macrophages via AMPK stimulation.


Subject(s)
Adenylate Kinase/physiology , Berberine/pharmacology , Inflammation Mediators/antagonists & inhibitors , Macrophages/drug effects , 3T3-L1 Cells , Adenylate Kinase/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/pathology , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Berberine/therapeutic use , Cells, Cultured , Drug Evaluation, Preclinical , Gene Expression Regulation/drug effects , Inflammation/drug therapy , Inflammation/genetics , Inflammation/pathology , Macrophages/enzymology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Mice, Transgenic , Receptors, Leptin/genetics
13.
Bioorg Med Chem ; 16(7): 3580-6, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18313308

ABSTRACT

Recent studies have shown that glucose-6-phosphate dehydrogenase (G6PD) is an effectual therapeutic target for metabolic disorders, including obesity and diabetes. In this study, we used in silico and conventional screening approaches to identify putative inhibitors of G6PD and found that gallated catechins (EGCG, GCG, ECG, CG), but not ungallated catechins (ECG, GC, EC, C), were NADP(+)-competitive inhibitors of G6PD and other enzymes that employ NADP(+) as a coenzyme, such as IDH and 6PGD.


Subject(s)
Catechin/analogs & derivatives , Coenzymes/metabolism , Glucosephosphate Dehydrogenase/antagonists & inhibitors , Glucosephosphate Dehydrogenase/metabolism , NADP/metabolism , 3T3-L1 Cells , Adipocytes/drug effects , Adipocytes/enzymology , Animals , Catechin/chemistry , Catechin/pharmacology , Humans , Kinetics , Mice , Molecular Structure , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...