Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Sci Immunol ; 8(88): eadg6155, 2023 10 13.
Article in English | MEDLINE | ID: mdl-37801517

ABSTRACT

High neonatal susceptibility to meningitis has been attributed to the anatomical barriers that act to protect the central nervous system (CNS) from infection being immature and not fully developed. However, the mechanisms by which pathogens breach CNS barriers are poorly understood. Using the Armstrong strain of lymphocytic choriomeningitis virus (LCMV) to study virus propagation into the CNS during systemic infection, we demonstrate that mortality in neonatal, but not adult, mice is high after infection. Virus propagated extensively from the perivenous sinus region of the dura mater to the leptomeninges, choroid plexus, and cerebral cortex. Although the structural barrier of CNS border tissues is comparable between neonates and adults, immunofluorescence staining and single-cell RNA sequencing analyses revealed that the neonatal dural immune cells are immature and predominantly composed of CD206hi macrophages, with major histocompatibility complex class II (MHCII)hi macrophages being rare. In adults, however, perivenous sinus immune cells were enriched in MHCIIhi macrophages that are specialized for producing antiviral molecules and chemokines compared with CD206hi macrophages and protected the CNS against systemic virus invasion. Our findings clarify how systemic pathogens enter the CNS through its border tissues and how the immune barrier at the perivenous sinus region of the dura blocks pathogen access to the CNS.


Subject(s)
Encephalitis, Viral , Lymphocytic Choriomeningitis , Meningitis, Viral , Meningoencephalitis , Mice , Animals , Central Nervous System , Meninges , Lymphocytic choriomeningitis virus
2.
Nat Commun ; 14(1): 2754, 2023 05 13.
Article in English | MEDLINE | ID: mdl-37179330

ABSTRACT

Active thermogenesis in the brown adipose tissue (BAT) facilitating the utilization of lipids and glucose is critical for maintaining body temperature and reducing metabolic diseases, whereas inactive BAT accumulates lipids in brown adipocytes (BAs), leading to BAT whitening. Although cellular crosstalk between endothelial cells (ECs) and adipocytes is essential for the transport and utilization of fatty acid in BAs, the angiocrine roles of ECs mediating this crosstalk remain poorly understood. Using single-nucleus RNA sequencing and knock-out male mice, we demonstrate that stem cell factor (SCF) derived from ECs upregulates gene expressions and protein levels of the enzymes for de novo lipogenesis, and promotes lipid accumulation by activating c-Kit in BAs. In the early phase of lipid accumulation induced by denervation or thermoneutrality, transiently expressed c-Kit on BAs increases the protein levels of the lipogenic enzymes via PI3K and AKT signaling. EC-specific SCF deletion and BA-specific c-Kit deletion attenuate the induction of the lipogenic enzymes and suppress the enlargement of lipid droplets in BAs after denervation or thermoneutrality in male mice. These data provide insight into SCF/c-Kit signaling as a regulator that promotes lipid accumulation through the increase of lipogenic enzymes in BAT when thermogenesis is inhibited.


Subject(s)
Adipocytes, Brown , Hypercholesterolemia , Animals , Male , Mice , Adipocytes, Brown/metabolism , Adipose Tissue, Brown/metabolism , Endothelial Cells/metabolism , Fatty Acids/metabolism , Hypercholesterolemia/metabolism , Lipogenesis/genetics , Mice, Knockout , Receptor Protein-Tyrosine Kinases/metabolism , Stem Cell Factor/genetics , Stem Cell Factor/metabolism , Thermogenesis/genetics , Proto-Oncogene Proteins c-kit
3.
J Am Soc Nephrol ; 34(5): 809-828, 2023 05 01.
Article in English | MEDLINE | ID: mdl-36724799

ABSTRACT

SIGNIFICANCE STATEMENT: Mesangial cells (MCs) in the kidney are essential to maintaining glomerular integrity, and their impairment leads to major glomerular diseases including diabetic nephropathy (DN). Although high blood glucose elicits abnormal alterations in MCs, the underlying mechanism is poorly understood. We show that YAP/TAZ are increased in MCs of patients with DN and two animal models of DN. High glucose directly induces activation of YAP/TAZ through the canonical Hippo pathway in cultured MCs. Hyperactivation of YAP/TAZ in mouse MCs recapitulates the hallmarks of DN. Activated YAP/TAZ bind and stabilize N-Myc, one of the Myc family. N-Myc stabilization leads to aberrant enhancement of its transcriptional activity and to MC impairments. Our findings shed light on how high blood glucose in diabetes mellitus leads to DN and support a rationale that lowering blood glucose in diabetes mellitus could delay DN pathogenesis. BACKGROUND: Mesangial cells (MCs) in the kidney are central to maintaining glomerular integrity, and their impairment leads to major glomerular diseases, including diabetic nephropathy (DN). Although high blood glucose elicits abnormal alterations in MCs, the underlying molecular mechanism is poorly understood. METHODS: Immunolocalization of YAP/TAZ and pathological features of PDGFRß + MCs were analyzed in the glomeruli of patients with DN, in Zucker diabetic fatty rats, and in Lats1/2i ΔPß mice. RiboTag bulk-RNA sequencing and transcriptomic analysis of gene expression profiles of the isolated MCs from control and Lats1/2iΔPß mice were performed. Immunoprecipitation analysis and protein stability of N-Myc were performed by the standard protocols. RESULTS: YAP and TAZ, the final effectors of the Hippo pathway, are highly increased in MCs of patients with DN and in Zucker diabetic fatty rats. Moreover, high glucose directly induces activation of YAP/TAZ through the canonical Hippo pathway in cultured MCs. Hyperactivation of YAP/TAZ in mouse model MCs recapitulates the hallmarks of DN, including excessive proliferation of MCs and extracellular matrix deposition, endothelial cell impairment, glomerular sclerosis, albuminuria, and reduced glomerular filtration rate. Mechanistically, activated YAP/TAZ bind and stabilize N-Myc protein, one of the Myc family of oncogenes. N-Myc stabilization leads to aberrant enhancement of its transcriptional activity and eventually to MC impairments and DN pathogenesis. CONCLUSIONS: Our findings shed light on how high blood glucose in diabetes mellitus leads to DN and support a rationale that lowering blood glucose in diabetes mellitus could delay DN pathogenesis.


Subject(s)
Diabetes Mellitus , Diabetic Nephropathies , Rats , Mice , Animals , Mesangial Cells/metabolism , Diabetic Nephropathies/metabolism , Blood Glucose/metabolism , Rats, Zucker , Protein Serine-Threonine Kinases/metabolism
4.
Article in English | MEDLINE | ID: mdl-33815558

ABSTRACT

OBJECTIVES: Ginsenoside Rg3 (Rg3), a main active component of Panax ginseng, has various therapeutic properties in literatures, and it has been studied for its potential use in obesity control due to its antiadipogenic effects in white adipocytes. However, little is known about its effects on brown adipocytes. METHODS: The mechanisms through which Rg3 inhibits differentiation, adipogenesis, and ER stress-mediated cell death in mouse primary brown adipocytes (MPBAs) are explored. RESULTS: Rg3 significantly induced cytotoxicity in differentiated MPBAs but not in undifferentiated MPBAs. Rg3 treatment downregulated the expression of differentiation and adipogenesis markers and the level of perilipin in MPBAs while upregulating the expression of lipolytic Kruppel-like factor genes. Rg3 also induced lipolysis and efflux of triglycerides from MPBAs and subsequently increased proinflammatory cytokine levels. Notably, Rg3 treatment resulted in elevation of ER stress and proapoptotic markers in MPBAs. CONCLUSIONS: Our results demonstrate that Rg3 is able to selectively exert cytotoxicity in differentiated MPBAs while leaving undifferentiated MPBAs intact, resulting in the induction of ER stress and subsequent cell death in MPBAs via regulation of various genes related to adipocyte differentiation, adipogenesis, lipolysis, and inflammation. These results indicate that further studies on the potential therapeutic applications of Rg3 are warranted.

5.
Nat Commun ; 11(1): 3866, 2020 07 31.
Article in English | MEDLINE | ID: mdl-32737287

ABSTRACT

Upon severe head injury (HI), blood vessels of the meninges and brain parenchyma are inevitably damaged. While limited vascular regeneration of the injured brain has been studied extensively, our understanding of meningeal vascular regeneration following head injury is quite limited. Here, we identify key pathways governing meningeal vascular regeneration following HI. Rapid and complete vascular regeneration in the meninges is predominantly driven by VEGFR2 signaling. Substantial increase of VEGFR2 is observed in both human patients and mouse models of HI, and endothelial cell-specific deletion of Vegfr2 in the latter inhibits meningeal vascular regeneration. We further identify the facilitating, stabilizing and arresting roles of Tie2, PDGFRß and Dll4 signaling, respectively, in meningeal vascular regeneration. Prolonged inhibition of this angiogenic process following HI compromises immunological and stromal integrity of the injured meninges. These findings establish a molecular framework for meningeal vascular regeneration after HI, and may guide development of wound healing therapeutics.


Subject(s)
Craniocerebral Trauma/genetics , Endothelial Cells/metabolism , Neovascularization, Physiologic/genetics , Regeneration/genetics , Signal Transduction/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Blood Vessels/metabolism , Blood Vessels/pathology , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cerebrovascular Circulation , Craniocerebral Trauma/metabolism , Craniocerebral Trauma/pathology , Disease Models, Animal , Endothelial Cells/pathology , Gene Expression Regulation/genetics , Humans , Macrophages/metabolism , Macrophages/pathology , Meninges/injuries , Meninges/metabolism , Mice , Mice, Knockout , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Wound Healing/genetics
6.
Nat Commun ; 11(1): 2980, 2020 06 12.
Article in English | MEDLINE | ID: mdl-32532986

ABSTRACT

Proper storage of excessive dietary fat into subcutaneous adipose tissue (SAT) prevents ectopic lipid deposition-induced insulin resistance, yet the underlying mechanism remains unclear. Here, we identify angiopoietin-2 (Angpt2)-integrin α5ß1 signaling as an inducer of fat uptake specifically in SAT. Adipocyte-specific deletion of Angpt2 markedly reduced fatty acid uptake and storage in SAT, leading to ectopic lipid accumulation in glucose-consuming organs including skeletal muscle and liver and to systemic insulin resistance. Mechanistically, Angpt2 activated integrin α5ß1 signaling in the endothelium and triggered fatty acid transport via CD36 and FATP3 into SAT. Genetic or pharmacological inhibition of the endothelial integrin α5ß1 recapitulated adipocyte-specific Angpt2 knockout phenotypes. Our findings demonstrate the critical roles of Angpt2-integrin α5ß1 signaling in SAT endothelium in regulating whole-body fat distribution for metabolic health and highlight adipocyte-endothelial crosstalk as a potential target for prevention of ectopic lipid deposition-induced lipotoxicity and insulin resistance.


Subject(s)
Angiopoietin-2/metabolism , Fatty Acids/metabolism , Insulin Resistance/physiology , Integrin alpha5beta1/metabolism , Lipid Metabolism/physiology , Subcutaneous Fat/metabolism , Adult , Angiopoietin-2/genetics , Animals , Cells, Cultured , Female , Gene Expression Profiling/methods , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Insulin Resistance/genetics , Integrin alpha5beta1/genetics , Lipid Metabolism/genetics , Lipids/analysis , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Middle Aged , Signal Transduction/genetics
7.
PLoS One ; 14(2): e0210854, 2019.
Article in English | MEDLINE | ID: mdl-30818368

ABSTRACT

Recent studies have demonstrated the immunomodulatory effects of heat-killed lactic acid bacteria. The aim of this study was to evaluate the protective effect of heat-killed Enterococcus faecalis EF-2001 (EF-2001) on a model of inflammatory bowel disease (IBD). A total of 28 female NC/Nga mice were divided into 4 treatment groups. Controls were fed a normal commercial diet. In the experimental groups, colitis was induced by rectal administration of dinitrobenzene sulfonic acid. Two groups were orally administered 2 and 17 mg/kg EF-2001, respectively. EF-2001 treatment decreased the expression of several cytokines, including cyclooxygenase (COX)-2, inducible nitric oxide synthase (iNOS), interferon (IFN)-γ, interleukin (IL)-1ß, and IL-6 in inflamed colon compared to the DNBS alone group. In addition, EF-2001 suppressed DNBS-induced colonic tissue destruction. Therefore, this study strongly suggests that EF-2001 could alleviate the inflammation associated with mouse IBD.


Subject(s)
Benzenesulfonates/toxicity , Colon/metabolism , Enterococcus faecalis , Inflammatory Bowel Diseases , Animals , Colon/pathology , Cyclooxygenase 2/metabolism , Cytokines/metabolism , Disease Models, Animal , Female , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Inflammatory Bowel Diseases/prevention & control , Mice
8.
J Exerc Rehabil ; 14(1): 16-23, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29511648

ABSTRACT

White adipose tissue (WAT) expansion is associated with angiogenesis. Although, activation of lipolysis by exercise induces adipocyte hypotrophy and reduction of fat mass, it is poorly understood whether exercise regulates angiogenesis by altering angiogenic gene expression in WAT. Therefore, the purpose of this study was to evaluate the effect of 6 weeks voluntary wheel running exercise on angiogenic gene expression in adipose tissues. Male C57BL/6J mice performed voluntary wheel running for 6 weeks. At 24 hr after the last exercise training, tibialis anterior (TA), soleus (Sol), epididymal WAT (eWAT), inguinal WAT (iWAT), and brown adipose tissue (BAT) were isolated and then the expressions of vascular endothelial growth factor A (VEGFA), angiopoietin1 (Ang1), Ang2, platelet-derived growth factor B (PDGF-B) and their corresponding receptors were analyzed by reverse transcription-polymerase chain reaction. In skeletal muscles, VEGFA expression was upregulated in TA and Sol and PDDGF-B expression was increased in Sol after exercise training. In eWAT, the expressions of VEGFA and Flk-1 were dramatically downregulated, whereas Ang2 and PDGFRß was upregulated after exercise training. In iWAT, VEGF expression was increased with the downregulation of Ang1. However, there was no alteration of any of these genes in BAT. These results suggest that angiogenic gene expression is altered by exercise training and voluntary wheel running regulates VEGFA, Ang1, and Ang2 expressions in a fat depot specific manner.

9.
Eur J Nutr ; 57(1): 391-403, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28439667

ABSTRACT

PURPOSE: Myricetin, a dietary flavonoid, is effective in the treatment of obesity and insulin resistance by increasing glucose transport and lipogenesis in adipocyte and diminishing systemic inflammation in obesity. However, it has not been revealed yet whether myricetin is associated with brown adipose tissue (BAT) activation that tightly mediates systemic energy metabolism. Therefore, this study assessed whether myricetin activated brown adipose tissue in db/db mouse. METHODS: Myricetin (400 mg/kg) in distilled water was fed daily by oral gavage to leptin receptor-deficient db/db male mice at 4 weeks of age for 14 weeks. Body weight change, glucose intolerance test, blood lipid profile and BAT activation using PET-CT were assessed. RESULTS: After myricetin treatment for 14 weeks, systemic insulin resistance and hepatic steatosis were significantly improved in db/db mice with body weight reduction and myricetin led to decreased adipocity, improved plasma lipid profiles and increased energy expenditure. Myricetin activated BAT by upregulating thermogenic protein expression and activating mitochondrial biogenesis, eventually increasing heat dissipation in skin after cold exposure. In iWAT, myricetin induced beige formation, increased thermogenic protein expression and activated mitochondrial biogenesis. Consistently, thermogenic gene expression was upregulated when myricetin was introduced in C3H10T1/2 cells during brown adipocytes differentiation. Moreover, the expression level of adiponectin was significantly increased in C3H10T1/2 cells, adipose tissues and plasma after myricetin treatment. CONCLUSIONS: These results highlight that myricetin prevents obesity and systemic insulin resistance by activating BAT and increasing adiponectin expression in BAT.


Subject(s)
Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/physiology , Flavonoids/administration & dosage , Insulin Resistance , Obesity/prevention & control , Adipocytes/drug effects , Adipocytes/metabolism , Adiponectin/genetics , Animals , Energy Metabolism/drug effects , Fatty Liver/prevention & control , Gene Expression/drug effects , Hyperlipidemias/prevention & control , Lipids/blood , Male , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/physiology , Receptors, Leptin/deficiency , Thermogenesis/drug effects , Thermogenesis/genetics , Weight Gain
10.
Liver Int ; 38(5): 803-812, 2018 05.
Article in English | MEDLINE | ID: mdl-28898508

ABSTRACT

BACKGROUND & AIMS: Hepatic steatosis is a common feature of patients with chronic hepatitis C. Previous reports have shown that the overexpression of hepatitis C virus core-encoding sequences (hepatitis C virus genotypes 3a and 1b) significantly induces intracellular triglyceride accumulation. However, the underlying mechanism has not yet been revealed. METHODS: To investigate whether Sirt1 is involved in hepatitis C virus-mediated hepatic steatosis, the overexpression of hepatitis C virus core 1b protein and Sirt1 and the knockdown of Sirt1 in HepG2 cells were performed. To confirm the results of the cellular experiment liver-specific Sirt1 KO mice with lentivirus-mediated hepatitis C virus core 1b overexpression were studied. RESULTS: Our results show that hepatitis C virus core 1b protein overexpression led to the accumulation of triglycerides in HepG2 cells. Notably the expression of PPARγ2 was dramatically increased at both the mRNA and protein levels by hepatitis C virus core 1b overexpression. The protein expression of Sirt1 is an upstream regulator of PPARγ2 and was also significantly increased after core 1b overexpression. In addition, the overexpression or knockdown of Sirt1 expression alone was sufficient to modulate p300-mediated PPARγ2 deacetylation. In vivo studies showed that hepatitis C virus core protein 1b-induced hepatic steatosis was attenuated in liver-specific Sirt1 KO mice by downregulation of PPARγ2 expression. CONCLUSIONS: Sirt1 mediates hepatitis C virus core protein 1b-induced hepatic steatosis by regulation of PPARγ2 expression.


Subject(s)
Fatty Liver/metabolism , Liver/pathology , Sirtuin 1/genetics , Viral Core Proteins/metabolism , Animals , Fatty Liver/genetics , Fatty Liver/virology , Gene Expression , Hep G2 Cells , Hepacivirus , Hepatitis C, Chronic/complications , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , PPAR gamma/metabolism , RNA, Messenger/metabolism , Triglycerides/metabolism
11.
Front Med ; 12(2): 130-138, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29119382

ABSTRACT

Brown adipose tissue (BAT) plays a fundamental role in maintaining body temperature by producing heat. BAT that had been know to exist only in mammals and the human neonate has received great attention for the treatment of obesity and diabetes due to its important function in energy metabolism, ever since it is recently reported that human adults have functional BAT. In addition, beige adipocytes, brown adipocytes in white adipose tissue (WAT), have also been shown to take part in whole body metabolism. Multiple lines of evidence demonstrated that transplantation or activation of BAT or/and beige adipocytes reversed obesity and improved insulin sensitivity. Furthermore, many genes involved in BATactivation and/or the recruitment of beige cells have been found, thereby providing new promising strategies for future clinical application of BAT activation to treat obesity and metabolic diseases. This review focuses on recent advances of BAT function in the metabolic aspect and the relationship between BAT and cancer cachexia, a pathological process accompanied with decreased body weight and increased energy expenditure in cancer patients. The underlying possible mechanisms to reduce BAT mass and its activity in the elderly are also discussed.


Subject(s)
Adipose Tissue, Brown/metabolism , Aging/metabolism , Cachexia/metabolism , Metabolic Syndrome/metabolism , Neoplasms/metabolism , Animals , Cachexia/pathology , Disease Models, Animal , Energy Metabolism , Humans , Neoplasms/pathology , Obesity/metabolism , Thermogenesis
12.
J Nutr Biochem ; 47: 21-28, 2017 09.
Article in English | MEDLINE | ID: mdl-28501702

ABSTRACT

Polycystic ovary syndrome (PCOS) is a complex endocrinopathy that is characterized by anovulation, hyperandrogenism and polycystic ovary. However, there is a lack of effective treatment for PCOS at present because the pathologic cause of PCOS has not been elucidated. Although it has been known that brown adipose tissue transplantation ameliorates PCOS by activating endogenous BAT, BAT transplantation is not applicable in clinic. Therefore, BAT activation with natural compound could be an effective treatment strategy for PCOS patients. Here, we found that 3 weeks of rutin (a novel compound for BAT activation) treatment increased BAT activation, thereby it improved thermogenesis and systemic insulin sensitivity in dehydroepiandrosterone (DHEA)-induced PCOS rat. In addition, the expression levels of ovarian steroidogenic enzymes such as P450C17, aromatase, 3ß-HSD, 17ß-HSD and STAR were up-regulated in rutin-treated PCOS rat. Furthermore, acyclicity and the serum level of luteinizing hormone were normalized, and a large number of mature ovulated follicle with a reduction of cystic formation were observed in PCOS rat after rutin treatment. Finally, rutin treatment surprisingly improved fertility and birth defect in PCOS rat. Collectively, our results indicate that rutin treatment significantly improves systemic insulin resistance and ovarian malfunction in PCOS, and our findings in this study provide a novel therapeutic option for the treatment of PCOS by activating BAT with rutin.


Subject(s)
Adipose Tissue, Brown/metabolism , Disease Models, Animal , Insulin Resistance , Ovary/physiopathology , Polycystic Ovary Syndrome/diet therapy , Rutin/therapeutic use , Thermogenesis , Adipose Tissue, Brown/pathology , Animals , Anovulation/etiology , Anovulation/prevention & control , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Anti-Obesity Agents/therapeutic use , Biomarkers/blood , Biomarkers/metabolism , Congenital Abnormalities/etiology , Congenital Abnormalities/prevention & control , Dehydroepiandrosterone , Enzyme Induction , Female , Infertility, Female/etiology , Infertility, Female/prevention & control , Luteinizing Hormone/antagonists & inhibitors , Luteinizing Hormone/blood , Ovary/metabolism , Ovary/pathology , Phosphoproteins/agonists , Phosphoproteins/genetics , Phosphoproteins/metabolism , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/pathology , Polycystic Ovary Syndrome/physiopathology , Rats, Sprague-Dawley , Thermography , Whole Body Imaging
13.
FASEB J ; 31(1): 333-345, 2017 01.
Article in English | MEDLINE | ID: mdl-28049156

ABSTRACT

Increasing energy expenditure through activation of brown adipose tissue (BAT) is a critical approach to treating obesity and diabetes. In this study, rutin, a natural compound extracted from mulberry and a drug used as a capillary stabilizer clinically for many years without any side effects, regulated whole-body energy metabolism by enhancing BAT activity. Rutin treatment significantly reduced adiposity, increased energy expenditure, and improved glucose homeostasis in both genetically obese (Db/Db) and diet-induced obesity (DIO) mice. Rutin also induced brown-like adipocyte (beige) formation in subcutaneous adipose tissue in both obesity mouse models. Mechanistically, we found that rutin directly bound to and stabilized SIRT1, leading to hypoacetylation of peroxisome proliferator-activated receptor γ coactivator-1α protein, which stimulated Tfam transactivation and eventually augmented the number of mitochondria and UCP1 activity in BAT. These findings reveal that rutin is a novel small molecule that activates BAT and may provide a novel therapeutic approach to the treatment of metabolic disorders.-Yuan, X., Wei, G., You, Y., Huang, Y., Lee, H. J., Dong, M., Lin, J., Hu, T., Zhang, H., Zhang, C., Zhou, H., Ye, R., Qi, X., Zhai, B., Huang, W., Liu, S., Xie, W., Liu, Q., Liu, X., Cui, C., Li, D., Zhan, J., Cheng, J., Yuan, Z., Jin, W. Rutin ameliorates obesity through brown fat activation.


Subject(s)
Adipose Tissue, Brown/physiology , Diet, High-Fat/adverse effects , Energy Metabolism/drug effects , Obesity/drug therapy , Rutin/pharmacology , Animals , Body Temperature Regulation/drug effects , Body Temperature Regulation/physiology , Cold Temperature , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Glucose Tolerance Test , HEK293 Cells , High Mobility Group Proteins/genetics , High Mobility Group Proteins/metabolism , Humans , Mice , Mice, Obese , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Sirtuin 1/genetics , Sirtuin 1/metabolism , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
14.
Sci Rep ; 6: 39193, 2016 12 16.
Article in English | MEDLINE | ID: mdl-27982067

ABSTRACT

Both brown adipocytes (BAC) and beige cells hold therapeutic potential for the treatment of metabolic disorders. Unfortunately, the amount and activity of these cells are limited in adults. Although BAC marker expression has been shown in peri-renal adipose tissues in children and adults, functional assessment is lacking. Furthermore, it is entirely unknown whether adipose progenitors are present in human embryo and able to give rise to BAC in situ during evolution. Therefore, adipose tissues in the interscapular and peri-renal regions were dissected from human embryo and subcutaneous white adipose tissues (sWAT) were obtained from an adult. After subjected to differentiation in vitro, adipocyte progenitors were detected present in all these adipose tissues. When stimulated for adipogenesis, differentiated adipocytes in the intercapular and peri-renal regions showed similar features: (1) induced BAC and beige cell marker expression including UCP1 and PRDM16 and comparable mitochondrion copy number; (2) similar gene expression patterns by RNA-Seq analysis; and (3) similar maximal oxygen consumption rates examined by respirometry. Nevertheless, stimulation of adipocyte progenitors in sWAT induces neither BAC and beige cell marker expression nor any change of oxygen consumption. In conclusion, peri-renal adipocyte progenitors in human embryo hold browning potential for BAC production.


Subject(s)
Adipocytes, Brown/metabolism , Embryo, Mammalian/cytology , Adipocytes, Brown/cytology , Adipocytes, White/cytology , Adipocytes, White/metabolism , Adipogenesis , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondria/metabolism , RNA/chemistry , RNA/isolation & purification , RNA/metabolism , Sequence Analysis, RNA , Stem Cells/cytology , Stem Cells/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptome , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
16.
J Lipid Res ; 57(6): 980-92, 2016 06.
Article in English | MEDLINE | ID: mdl-27044658

ABSTRACT

Many types of apoptotic cells are phagocytosed and digested by macrophages. Adipocytes can be hundreds of times larger than macrophages, so they are too large to be digested by conventional phagocytic processes. The nature of the interaction between macrophages and apoptotic adipocytes has not been studied in detail. We describe a cellular process, termed exophagy, that is important for macrophage clearance of dead adipocytes and adipose tissue homeostasis. Using mouse models of obesity, human tissue, and a cell culture model, we show that macrophages form hydrolytic extracellular compartments at points of contact with dead adipocytes using local actin polymerization. These compartments are acidic and contain lysosomal enzymes delivered by exocytosis. Uptake and complete degradation of adipocyte fragments, which are released by extracellular hydrolysis, leads to macrophage foam cell formation. Exophagy-mediated foam cell formation is a highly efficient means by which macrophages internalize large amounts of lipid, which may ultimately overwhelm the metabolic capacity of the macrophage. This process provides a mechanism for degradation of objects, such as dead adipocytes, that are too large to be phagocytosed by macrophages.


Subject(s)
Adipocytes/physiology , Exocytosis/physiology , Macrophages/physiology , Obesity/physiopathology , Phagocytosis/physiology , Adipose Tissue/metabolism , Adipose Tissue/physiology , Animals , Foam Cells/pathology , Foam Cells/physiology , Humans , Lysosomes/physiology , Macrophages/metabolism , Mice , Obesity/metabolism
17.
Proc Natl Acad Sci U S A ; 113(10): 2708-13, 2016 Mar 08.
Article in English | MEDLINE | ID: mdl-26903641

ABSTRACT

Polycystic ovary syndrome (PCOS), which is characterized by anovulation, hyperandrogenism, and polycystic ovaries, is a complex endocrinopathy. Because the cause of PCOS at the molecular level is largely unknown, there is no cure or specific treatment for PCOS. Here, we show that transplantation of brown adipose tissue (BAT) reversed anovulation, hyperandrogenism, and polycystic ovaries in a dehydroepiandrosterone (DHEA)-induced PCOS rat. BAT transplantation into a PCOS rat significantly stabilized menstrual irregularity and improved systemic insulin sensitivity up to a normal level, which was not shown in a sham-operated or muscle-transplanted PCOS rat. Moreover, BAT transplantation, not sham operation or muscle transplantation, surprisingly improved fertility in PCOS rats. Interestingly, BAT transplantation activated endogenous BAT and thereby increased the circulating level of adiponectin, which plays a prominent role in whole-body energy metabolism and ovarian physiology. Consistent with BAT transplantation, administration of adiponectin protein dramatically rescued DHEA-induced PCOS phenotypes. These results highlight that endogenous BAT activity is closely related to the development of PCOS phenotypes and that BAT activation might be a promising therapeutic option for the treatment of PCOS.


Subject(s)
Adipose Tissue, Brown/transplantation , Infertility, Female/surgery , Insulin Resistance , Polycystic Ovary Syndrome/surgery , Adiponectin/blood , Adiponectin/pharmacology , Adult , Analysis of Variance , Animals , Blood Glucose/metabolism , Dehydroepiandrosterone , Energy Metabolism/drug effects , Estrous Cycle/drug effects , Female , Humans , Insulin/blood , Male , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/metabolism , Rats, Sprague-Dawley , Treatment Outcome
18.
Food Funct ; 6(2): 401-8, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25554947

ABSTRACT

Mulberry extract (ME) has been shown to possess beneficial effects towards obesity, but its mechanism is still unclear. In small mammals, mitochondria enriched brown adipose tissue (BAT) is known to convert protein's electrochemical energy to heat and maintain a constant body temperature. Improving the mitochondrial function or increasing the number of mitochondria could promote the metabolism of carbohydrate and fat. Thus, this study was designed to investigate the mitochondrial function regulated by ME and mulberry wine extract (MWE) during the brown adipogenesis. The C3H10T1/2 mesenchymal stem cell was treated with ME and MWE, both of which significantly (p < 0.05) increased the expression levels of fatty acid oxidation related genes such as peroxisome proliferator-activated receptor-γ coactivator-1α, PR domain-containing 16 and carnitine palmitoyltransferase 1α during brown adipogenesis. These changes were accompanied with increases in mitochondrial oxidative complex proteins upon ME and/or MWE exposure. Notably, ME and/or MWE also significantly (p < 0.05) increased the expression of the transcription factor A and the nuclear respiratory factor-1, which are the key transcription factors of mitochondrial biogenesis. In parallel, the mitochondrial copy number and brown adipose tissue specific gene-uncoupling protein-1 expression were dramatically (p < 0.05) elevated after ME or MWE treatment. Cyanidin-3-glucoside (Cy-3-glu) was found to be one of the most abundant anthocyanins in ME and MWE. Therefore, the BAT regulatory activity of ME and MWE might be, at least in part, due to the effect of Cy-3-glu. These results suggested that ME and MWE could ameliorate metabolic disease through an improvement in mitochondrial functions.


Subject(s)
Adipogenesis/drug effects , Adipose Tissue, Brown/metabolism , Mitochondria/drug effects , Morus/chemistry , Plant Extracts/pharmacology , Wine/analysis , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/drug effects , Animals , Anthocyanins/pharmacology , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Glucosides/pharmacology , Ion Channels/genetics , Ion Channels/metabolism , Lipid Metabolism/drug effects , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C3H , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Nuclear Respiratory Factor 1/genetics , Nuclear Respiratory Factor 1/metabolism , Organelle Biogenesis , Oxygen Consumption/drug effects , PPAR gamma/genetics , PPAR gamma/metabolism , Phosphorylation , Transcription Factors/genetics , Transcription Factors/metabolism , Uncoupling Protein 1 , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
19.
J Clin Invest ; 117(12): 3684-95, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18060029

ABSTRACT

Little is known about whether bone marrow-derived circulating progenitor cells (BMDCPCs) can transdifferentiate into adipocytes in adipose tissues or play a role in expanding adipocyte number during adipose tissue growth. Using a mouse bone marrow transplantation model, we addressed whether BMDCPCs can transdifferentiate into adipocytes under standard conditions as well as in the settings of diet-induced obesity, rosiglitazone treatment, and exposure to G-CSF. We also addressed the possibility of transdifferentiation to adipocytes in a murine parabiosis model. In each of these settings, our findings indicated that BMDCPCs did not transdifferentiate into either unilocular or multilocular adipocytes in adipose tissues. Most BMDCPCs became resident and phagocytic macrophages in adipose tissues--which resembled transdifferentiated multilocular adipocytes by appearance, but displayed cell surface markers characteristic for macrophages - in the absence of adipocyte marker expression. When exposed to adipogenic medium in vitro, bone marrow cells differentiated into multilocular, but not unilocular, adipocytes, but transdifferentiation was not observed in vivo, even in the contexts of adipose tissue regrowth or dermal wound healing. Our results suggest that BMDCPCs do not transdifferentiate into adipocytes in vivo and play little, if any, role in expanding the number of adipocytes during the growth of adipose tissues.


Subject(s)
Adipocytes/physiology , Adipose Tissue/physiology , Antigens, Differentiation/biosynthesis , Bone Marrow Cells/physiology , Cell Differentiation/physiology , Macrophages/physiology , Stem Cells/physiology , Adipocytes/cytology , Adipose Tissue/cytology , Animals , Bone Marrow Cells/cytology , Bone Marrow Transplantation , Diet/adverse effects , Gene Expression Profiling , Gene Expression Regulation/physiology , Macrophages/cytology , Mice , Mice, Transgenic , Obesity/chemically induced , Obesity/metabolism , Obesity/pathology , Oligonucleotide Array Sequence Analysis , Organ Specificity/physiology , Parabiosis , Stem Cells/cytology
20.
FASEB J ; 18(11): 1200-8, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15284220

ABSTRACT

The angiopoietin (Ang) family of growth factors includes Ang1, Ang2, Ang3, and Ang4, all of which bind to the endothelial receptor tyrosine kinase Tie2. Ang3 (mouse) and Ang4 (human) are interspecies orthologs. In experiments with human endothelial cell lines, Ang3 was identified as an antagonist of Tie2 and Ang4 was identified as an agonist of Tie2. However, the biological roles of Ang3 and Ang4 are unknown. We examined the biological effect of recombinant Ang3 and Ang4 proteins in primary cultured endothelial cells and in vivo in mice. Recombinant Ang3 and Ang4 formed disulfide-linked dimers. Ang4 (400 ng/mL) markedly increased Tie2 and Akt phosphorylation in primary cultured HUVECs whereas Ang3 (400 ng/mL) did not produce significant changes. Accordingly, Ang4, but not Ang3, induced survival and migration in primary cultured HUVECs. Unexpectedly, intravenously administered Ang3 (30 microg) was more potent than Ang4 (30 microg) in phosphorylating the Tie2 receptor in lung tissue from mice in vivo. Accordingly, Ang3 was more potent than Ang4 in phosphorylating Akt in primary cultured mouse lung microvascular endothelial cells. Ang3 and Ang4 both produced potent corneal angiogenesis extending from the limbus across the mouse cornea in vivo. Thus, Ang3 and Ang4 are agonists of Tie2, but mouse Ang3 has strong activity only on endothelial cells of its own species.


Subject(s)
Angiopoietins/physiology , Corneal Neovascularization/etiology , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Receptor, TIE-2/agonists , Ribonucleases/physiology , Angiopoietin-1/chemistry , Angiopoietin-1/genetics , Angiopoietin-1/pharmacology , Angiopoietin-1/physiology , Angiopoietin-2/chemistry , Angiopoietin-2/genetics , Angiopoietin-2/pharmacology , Angiopoietin-2/physiology , Angiopoietin-Like Protein 1 , Angiopoietin-Like Protein 4 , Angiopoietin-like Proteins , Angiopoietins/chemistry , Angiopoietins/genetics , Angiopoietins/pharmacology , Animals , Apoptosis/drug effects , Blood Proteins/genetics , Blood Proteins/physiology , Capillaries/cytology , Cell Movement/drug effects , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Dimerization , Endothelial Cells/drug effects , Endothelium, Vascular/cytology , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/pharmacology , Intercellular Signaling Peptides and Proteins/physiology , Lung/blood supply , Male , Mice , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Recombinant Fusion Proteins/physiology , Ribonucleases/chemistry , Ribonucleases/genetics , Ribonucleases/pharmacology , Species Specificity , Transfection , Umbilical Veins
SELECTION OF CITATIONS
SEARCH DETAIL
...