Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 150
Filter
Add more filters

Publication year range
1.
Cell ; 161(7): 1576-1591, 2015 Jun 18.
Article in English | MEDLINE | ID: mdl-26091038

ABSTRACT

The synthesis of type I collagen, the main component of bone matrix, precedes the expression of Runx2, the earliest determinant of osteoblast differentiation. We hypothesized that the energetic needs of osteoblasts might explain this apparent paradox. We show here that glucose, the main nutrient of osteoblasts, is transported in these cells through Glut1, whose expression precedes that of Runx2. Glucose uptake favors osteoblast differentiation by suppressing the AMPK-dependent proteasomal degradation of Runx2 and promotes bone formation by inhibiting another function of AMPK. While RUNX2 cannot induce osteoblast differentiation when glucose uptake is compromised, raising blood glucose levels restores collagen synthesis in Runx2-null osteoblasts and initiates bone formation in Runx2-deficient embryos. Moreover, RUNX2 favors Glut1 expression, and this feedforward regulation between RUNX2 and Glut1 determines the onset of osteoblast differentiation during development and the extent of bone formation throughout life. These results reveal an unexpected intricacy between bone and glucose metabolism.


Subject(s)
Cell Differentiation , Core Binding Factor Alpha 1 Subunit/metabolism , Glucose/metabolism , Osteoblasts/metabolism , Osteogenesis , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/genetics , Amino Acid Sequence , Animals , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Glucose Transporter Type 1/metabolism , Homeostasis , Mice , Osteoblasts/cytology , Sequence Alignment , Skull/cytology
2.
Cell ; 159(4): 709-13, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25417146

ABSTRACT

Mammalian aging can be delayed with genetic, dietary, and pharmacologic approaches. Given that the elderly population is dramatically increasing and that aging is the greatest risk factor for a majority of chronic diseases driving both morbidity and mortality, it is critical to expand geroscience research directed at extending human healthspan.


Subject(s)
Aging/physiology , Chronic Disease , Aging/pathology , Animals , Biomedical Research , Epigenesis, Genetic , Gene-Environment Interaction , Humans
3.
Nature ; 543(7645): 385-390, 2017 03 16.
Article in English | MEDLINE | ID: mdl-28273060

ABSTRACT

Bone has recently emerged as a pleiotropic endocrine organ that secretes at least two hormones, FGF23 and osteocalcin, which regulate kidney function and glucose homeostasis, respectively. These findings have raised the question of whether other bone-derived hormones exist and what their potential functions are. Here we identify, through molecular and genetic analyses in mice, lipocalin 2 (LCN2) as an osteoblast-enriched, secreted protein. Loss- and gain-of-function experiments in mice demonstrate that osteoblast-derived LCN2 maintains glucose homeostasis by inducing insulin secretion and improves glucose tolerance and insulin sensitivity. In addition, osteoblast-derived LCN2 inhibits food intake. LCN2 crosses the blood-brain barrier, binds to the melanocortin 4 receptor (MC4R) in the paraventricular and ventromedial neurons of the hypothalamus and activates an MC4R-dependent anorexigenic (appetite-suppressing) pathway. These results identify LCN2 as a bone-derived hormone with metabolic regulatory effects, which suppresses appetite in a MC4R-dependent manner, and show that the control of appetite is an endocrine function of bone.


Subject(s)
Appetite Regulation/physiology , Bone and Bones/metabolism , Lipocalin-2/metabolism , Receptor, Melanocortin, Type 4/metabolism , Animals , Blood-Brain Barrier/metabolism , Bone and Bones/cytology , Cyclic AMP/metabolism , Eating/physiology , Female , Fibroblast Growth Factor-23 , Glucose/metabolism , Homeostasis , Hypothalamus/cytology , Hypothalamus/metabolism , Insulin/metabolism , Insulin Resistance , Insulin Secretion , Male , Mice , Neurons/metabolism , Obesity/metabolism , Osteoblasts/metabolism , Paraventricular Hypothalamic Nucleus/cytology , Thinness/metabolism
5.
Physiol Genomics ; 53(11): 456-472, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34643091

ABSTRACT

Excessive long-term consumption of dietary carbohydrates, including glucose, sucrose, or fructose, has been shown to have significant impact on genome-wide gene expression, which likely results from changes in metabolic substrate flux. However, there has been no comprehensive study on the acute effects of individual sugars on the genome-wide gene expression that may reveal the genetic changes altering signaling pathways, subsequent metabolic processes, and ultimately physiological/pathological responses. Considering that gene expressions in response to acute carbohydrate ingestion might be different in nutrient sensitive and insensitive mammals, we conducted comparative studies of genome-wide gene expression by deep mRNA sequencing of the liver in nutrient sensitive C57BL/6J and nutrient insensitive BALB/cJ mice. Furthermore, to determine the temporal responses, we compared livers from mice in the fasted state and following ingestion of standard laboratory mouse chow supplemented with plain drinking water or water containing 20% glucose, sucrose, or fructose. Supplementation with these carbohydrates induced unique extents and temporal changes in gene expressions in a strain specific manner. Fructose and sucrose stimulated gene changes peaked at 3 h postprandial, whereas glucose effects peaked at 12 h and 6 h postprandial in C57BL/6J and BABL/cJ mice, respectively. Network analyses revealed that fructose changed genes were primarily involved in lipid metabolism and were more complex in C57BL/6J than in BALB/cJ mice. These data demonstrate that there are qualitative and antitative differences in the normal physiological responses of the liver between these two strains of mice and C57BL/6J is more sensitive to sugar intake than BALB/cJ.


Subject(s)
Dietary Carbohydrates/administration & dosage , Dietary Supplements , Liver/metabolism , Transcriptome/drug effects , Transcriptome/genetics , Animals , Dietary Carbohydrates/metabolism , Down-Regulation/drug effects , Down-Regulation/genetics , Eating , Fasting , Fructose/administration & dosage , Fructose/metabolism , Glucose/administration & dosage , Glucose/metabolism , Lipid Metabolism/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Signal Transduction/genetics , Species Specificity , Sucrose/administration & dosage , Sucrose/metabolism , Up-Regulation/drug effects , Up-Regulation/genetics
6.
J Biol Chem ; 295(44): 15045-15053, 2020 10 30.
Article in English | MEDLINE | ID: mdl-32848017

ABSTRACT

Previously we reported that adipocyte SNAP23 (synaptosome-associated protein of 23 kDa) deficiency blocks the activation of macroautophagy, leading to an increased abundance of BAX, a pro-death Bcl-2 family member, and activation and adipocyte cell death both in vitro and in vivo Here, we found that knockdown of SNAP23 inhibited the association of the autophagosome regulators ATG16L1 and ATG9 compartments by nutrient depletion and reduced the formation of ATG16L1 membrane puncta. ATG16L1 knockdown inhibited autophagy flux and increased BAX protein levels by suppressing BAX degradation. The elevation in BAX protein had no effect on BAX activation or cell death in the nutrient-replete state. However, following nutrient depletion, BAX was activated with a concomitant induction of cell death. Co-immunoprecipitation analyses demonstrated that SNAP23 and ATG16L1 proteins form a stable complex independent of nutrient condition, whereas in the nutrient-depleted state, BAX binds to SNAP23 to form a ternary BAX-SNAP23-ATG16L1 protein complex. Taken together, these data support a model in which SNAP23 plays a crucial function as a scaffold for ATG16L1 necessary for the suppression of BAX activation and induction of the intrinsic cell death program.


Subject(s)
Apoptosis/physiology , Autophagy-Related Proteins/physiology , Autophagy/physiology , bcl-2-Associated X Protein/metabolism , Animals , Autophagy-Related Proteins/metabolism , Mice , NIH 3T3 Cells , Protein Binding , Qb-SNARE Proteins/genetics , Qb-SNARE Proteins/metabolism , Qc-SNARE Proteins/genetics , Qc-SNARE Proteins/metabolism , Subcellular Fractions/metabolism
7.
J Biol Chem ; 295(15): 4809-4821, 2020 04 10.
Article in English | MEDLINE | ID: mdl-32075912

ABSTRACT

The liver maintains metabolic homeostasis by integrating the regulation of nutrient status with both hormonal and neural signals. Many studies on hepatic signaling in response to nutrients have been conducted in mice. However, no in-depth study is currently available that has investigated genome-wide changes in gene expression during the normal physiological fasting-feeding cycle in nutrient-sensitive and -insensitive mice. Using two strains of mice, C57BL/6J and BALB/cJ, and deploying deep RNA-Seq complemented with quantitative RT-PCR, we found that feeding causes substantial and transient changes in gene expression in the livers of both mouse strains. The majority of significantly changed transcripts fell within the areas of biological regulation and cellular and metabolic processes. Among the metabolisms of three major types of macronutrients (i.e. carbohydrates, proteins, and lipids), feeding affected lipid metabolism the most. We also noted that the C57BL/6J and BALB/cJ mice significantly differed in gene expression and in changes in gene expression in response to feeding. In both fasted and fed states, both mouse strains shared common expression patterns for about 10,200 genes, and an additional 400-600 genes were differentially regulated in one strain but not the other. Among the shared genes, more lipogenic genes were induced upon feeding in BABL/cJ than in C57BL/6J mice. In contrast, in the population of differentially enriched genes, C57BL/6J mice expressed more genes involved in lipid metabolism than BALB/cJ mice. In summary, these results reveal that the two mouse strains used here exhibit several differences in feeding-induced hepatic responses in gene expression, especially in lipogenic genes.


Subject(s)
Biomarkers/metabolism , Eating , Fasting , Gene Expression Regulation , Lipid Metabolism , Liver/metabolism , Animals , Gene Expression Profiling , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Species Specificity
8.
J Biol Chem ; 294(23): 9076-9083, 2019 06 07.
Article in English | MEDLINE | ID: mdl-31028171

ABSTRACT

The Mediator complex plays a critical role in the regulation of transcription by linking transcription factors to RNA polymerase II. By examining mouse livers, we have found that in the fasted state, the Mediator complex exists primarily as an approximately 1.2-MDa complex, consistent with the size of the large Mediator complex, whereas following feeding, it converts to an approximately 600-kDa complex, consistent with the size of the core Mediator complex. This dynamic change is due to the dissociation and degradation of the kinase module that includes the MED13, MED12, cyclin-dependent kinase 8 (CDK8), and cyclin C (CCNC) subunits. The dissociation and degradation of the kinase module are dependent upon nutrient activation of mTORC1 that is necessary for the induction of lipogenic gene expression because pharmacological or genetic inhibition of mTORC1 in the fed state restores the kinase module. The degradation but not dissociation of the kinase module depends upon the E3 ligase, SCFFBW7 In addition, genetically insulin-resistant and obese db/db mice in the fasted state displayed elevated lipogenic gene expression and loss of the kinase module that was reversed following mTORC1 inhibition. These data demonstrate that the assembly state of the Mediator complex undergoes physiologic regulation during normal cycles of fasting and feeding in the mouse liver. Furthermore, the assembly state of the Mediator complex is dysregulated in states of obesity and insulin resistance.


Subject(s)
Insulin Resistance , Mediator Complex/metabolism , Obesity/pathology , Animals , Cell Nucleus/metabolism , Cyclin C/metabolism , Cyclin-Dependent Kinase 8/metabolism , Liver/metabolism , Male , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Nutrients/administration & dosage , Obesity/metabolism , Protein Subunits/metabolism , SKP Cullin F-Box Protein Ligases/deficiency , SKP Cullin F-Box Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/metabolism , Signal Transduction/drug effects , Sirolimus/pharmacology
9.
Int J Mol Sci ; 21(13)2020 Jul 05.
Article in English | MEDLINE | ID: mdl-32635651

ABSTRACT

Adipose tissue is an important regulator of whole-body metabolism and energy homeostasis. The unprecedented growth of obesity and metabolic disease worldwide has required paralleled advancements in research on this dynamic endocrine organ system. Single-cell RNA sequencing (scRNA-seq), a highly meticulous methodology used to dissect tissue heterogeneity through the transcriptional characterization of individual cells, is responsible for facilitating critical advancements in this area. The unique investigative capabilities achieved by the combination of nanotechnology, molecular biology, and informatics are expanding our understanding of adipose tissue's composition and compartmentalized functional specialization, which underlie physiologic and pathogenic states, including adaptive thermogenesis, adipose tissue aging, and obesity. In this review, we will summarize the use of scRNA-seq and single-nuclei RNA-seq (snRNA-seq) in adipocyte biology and their applications to obesity and diabetes research in the hopes of increasing awareness of the capabilities of this technology and acting as a catalyst for its expanded use in further investigation.


Subject(s)
Adipocytes, Beige/metabolism , Adipose Tissue/metabolism , Genomics , Single-Cell Analysis , Adipose Tissue/immunology , Animals , Cells, Cultured , Humans , Obesity/immunology , Sequence Analysis, RNA , Stem Cells/physiology , Transcriptome
10.
Molecules ; 25(12)2020 Jun 18.
Article in English | MEDLINE | ID: mdl-32570757

ABSTRACT

The family with sequence similarity 83 (FAM83) protein family G (FAM83G) possesses a predicted consensus phosphorylation motif for serine/threonine-protein kinase D1/protein kinase C mu (PKD1/PKCµ) at serine residue 356 (S356). In this study, overexpressed wild-type FAM83G coimmunoprecipitated with PKD1/PKCµ in Chinese hamster ovary (CHO) cells inhibited heat shock protein 27 (HSP27) phosphorylation at S82 and reduced the living cell number. The expression of a FAM83G phosphorylation-resistant mutant (S356A-FAM83G) had no effect on the living cell number or the induction of spontaneous apoptosis. By contrast, the introduction of a synthetic peptide encompassing FAM83G S356 into HCT116 and HepG2 cells decreased HSP27 S15 and S82 phosphorylation and induced spontaneous apoptosis. On the other hand, the introduction of FAM83G phosphorylation-resistant mutant synthesized peptides (S356A-AF-956 and S356A-AG-066) did not reduce the living cell number or induce spontaneous apoptosis. The endogenous expression of HSP27 and FAM83G was apparently greater in HCT116 and HepG2 cells compared with in CHO cells. In various types of lung cancer cell lines, the FAM83G messenger RNA (mRNA) level in non-small lung cancer cells was at a similar level to that in non-cancerous cells. However, the FAM83G mRNA level in the small cell lung cancer cell lines was variable, and the HSP27 mRNA level in FAM83G mRNA-rich types was greater than that in FAM83G mRNA-normal range types. Taken together, these data demonstrate that FAM83G S356 phosphorylation modulates HSP27 phosphorylation and apoptosis regulation and that HSP27 is a counterpart of FAM83G.


Subject(s)
Apoptosis , Carcinoma, Non-Small-Cell Lung/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lung Neoplasms/metabolism , Neoplasm Proteins/metabolism , Alcohol Oxidoreductases , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , HCT116 Cells , Hep G2 Cells , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Peptides/chemical synthesis , Peptides/chemistry , Peptides/pharmacology , Phosphorylation
11.
Molecules ; 25(3)2020 Jan 23.
Article in English | MEDLINE | ID: mdl-31979355

ABSTRACT

Dapagliflozin, empagliflozin, tofogliflozin, selective inhibitors of sodium-glucose cotransporter 2 (SGLT2), is used clinically to reduce circulation glucose levels in patients with type 2 diabetes mellitus by blocking the reabsorption of glucose by the kidneys. Dapagliflozin is metabolized and inactivated by UGT1A9. Empagliflozin is metabolized and inactivated by UGT1A9 and by other related isoforms UGT2B7, UGT1A3, and UGT1A8. Tofogliflozin is metabolized and inactivated by five different enzymes CYP2C18, CYP3A4, CYP3A5, CYP4A11, and CYP4F3. Dapagliflozin treatment of HCT116 cells, which express SGLT2 but not UGT1A9, results in the loss of cell adhesion, whereas HepG2 cells, which express both SGLT2 and UGT1A9, are resistant to the adhesion-related effects of dapagliflozin. PANC-1 and H1792 cells, which do not express either SGLT2 or UGT1A9, are also resistant to adhesion related effects of dapagliflozin. On the other hand, either empagliflozin or tofogliflozin treatment of HCT116, HepG2, PANC-1, and H1792 cells are resistant to the adhesion-related effects as observed in dapagliflozin treated HCT116 cells. Knockdown of UGT1A9 by shRNA in HepG2 cells increased dapagliflozin sensitivity, whereas the overexpression of UGT1A9 in HCT116 cells protected against dapagliflozin-dependent loos of cell adhesion. Dapagliflozin treatment had no effect on cellular interactions with fibronectin, vitronectin, or laminin, but it induced a loss of interaction with collagen I and IV. In parallel, dapagliflozin treatment reduced protein levels of the full-length discoidin domain receptor I (DDR1), concomitant with appearance of DDR1 cleavage products and ectodomain shedding of DDR1. In line with these observations, unmetabolized dapagliflozin increased ADAM10 activity. Dapagliflozin treatment also significantly reduced Y792 tyrosine phosphorylation of DDR1 leading to decrement of DDR1 function and detachment of cancer cells. Concomitant with these lines of results, we experienced that CEA in patients with colon cancer, which express SGLT2 but not UGT1A9, and type 2 diabetes mellitus treated by dapagliflozin in addition to chemotherapy was decreased (case 1). CEA in patients with colon cancer, which express SGLT2 but not UGT1A9, and type 2 diabetes mellitus was treated by dapagliflozin alone after radiation therapy was decreased but started to rise after cessation of dapagliflozin (case 2). CA19-9 in two of patients with pancreatic cancer and type 2 diabetes mellitus was resistant to the combination therapy of dapagliflozin and chemotherapy (case 3 and 4 respectively). PIVKAII in patients with liver cancer and type 2 diabetes mellitus, and CYFRA in patients with squamous lung cancer and type 2 diabetes mellitus was also resistant the combination therapy of dapagliflozin and chemotherapy (case 5 and 6 respectively). Taken together, these data suggest a potential role for dapagliflozin anticancer therapy against colon cancer cells that express SGLT2, but not UGT1A9.


Subject(s)
ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/metabolism , Antineoplastic Agents/pharmacology , Benzhydryl Compounds/pharmacology , Cell Adhesion/drug effects , Discoidin Domain Receptor 1/metabolism , Glucosides/pharmacology , Membrane Proteins/metabolism , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Carcinoembryonic Antigen/metabolism , Cell Line, Tumor , Collagen Type I/metabolism , Collagen Type IV/metabolism , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Fibronectins/metabolism , Gene Knockdown Techniques , Glucuronosyltransferase/metabolism , Humans , Laminin/metabolism , Phosphorylation , RNA, Small Interfering , Vitronectin/metabolism
12.
J Biol Chem ; 293(20): 7578-7591, 2018 05 18.
Article in English | MEDLINE | ID: mdl-29650758

ABSTRACT

The systems integration of whole-body metabolism and immune signaling are central homeostatic mechanisms necessary for maintenance of normal physiology, and dysregulation of these processes leads to a variety of chronic disorders. However, the intracellular mechanisms responsible for cell-autonomous cross-talk between the inflammatory signaling pathways and metabolic flux have remained enigmatic. In this study, we discovered that the fructose-2,6-bisphosphatase TIGAR (Tp53-induced glycolysis and apoptosis regulator) critically regulates NF-κB activation. We found that TIGAR potently inhibits NF-κB-dependent gene expression by suppressing the upstream activation of IKKß phosphorylation and kinase activation. This inhibition occurred through a direct binding competition between NEMO and TIGAR for association with the linear ubiquitination assembly complex (LUBAC). This competition prevented linear ubiquitination of NEMO, which is required for activation of IKKß and other downstream targets. Furthermore, a TIGAR phosphatase activity-deficient mutant was equally effective as WT TIGAR in inhibiting NEMO linear ubiquitination, IKKß phosphorylation/activation, and NF-κB signaling, indicating that TIGAR's effect on NF-κB signaling is due to its interaction with LUBAC. Physiologically, TIGAR knockout mice displayed enhanced adipose tissue NF-κB signaling, whereas adipocyte-specific overexpression of TIGAR suppressed adipose tissue NF-κB signaling. Together, these results demonstrate that TIGAR has a nonenzymatic molecular function that modulates the NF-κB signaling pathway by directly inhibiting the E3 ligase activity of LUBAC.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Multiprotein Complexes/antagonists & inhibitors , NF-kappa B/metabolism , Proteins/physiology , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin/metabolism , 3T3-L1 Cells , Animals , Apoptosis Regulatory Proteins , Gene Expression Regulation , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , NF-kappa B/genetics , Phosphoric Monoester Hydrolases , Phosphorylation , Ubiquitination
13.
J Biol Chem ; 292(21): 8918-8932, 2017 05 26.
Article in English | MEDLINE | ID: mdl-28351837

ABSTRACT

Brown adipose tissue is important for maintaining energy homeostasis and adaptive thermogenesis in rodents and humans. As disorders arising from dysregulated energy metabolism, such as obesity and metabolic diseases, have increased, so has interest in the molecular mechanisms of adipocyte biology. Using a functional screen, we identified cyclin C (CycC), a conserved subunit of the Mediator complex, as a novel regulator for brown adipocyte formation. siRNA-mediated CycC knockdown (KD) in brown preadipocytes impaired the early transcriptional program of differentiation, and genetic KO of CycC completely blocked the differentiation process. RNA sequencing analyses of CycC-KD revealed a critical role of CycC in activating genes co-regulated by peroxisome proliferator activated receptor γ (PPARγ) and CCAAT/enhancer-binding protein α (C/EBPα). Overexpression of PPARγ2 or addition of the PPARγ ligand rosiglitazone rescued the defects in CycC-KO brown preadipocytes and efficiently activated the PPARγ-responsive promoters in both WT and CycC-KO cells, suggesting that CycC is not essential for PPARγ transcriptional activity. In contrast, CycC-KO significantly reduced C/EBPα-dependent gene expression. Unlike for PPARγ, overexpression of C/EBPα could not induce C/EBPα target gene expression in CycC-KO cells or rescue the CycC-KO defects in brown adipogenesis, suggesting that CycC is essential for C/EBPα-mediated gene activation. CycC physically interacted with C/EBPα, and this interaction was required for C/EBPα transactivation domain activity. Consistent with the role of C/EBPα in white adipogenesis, CycC-KD also inhibited differentiation of 3T3-L1 cells into white adipocytes. Together, these data indicate that CycC activates adipogenesis in part by stimulating the transcriptional activity of C/EBPα.


Subject(s)
Adipocytes, Brown/metabolism , Adipogenesis , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Differentiation , Cyclin C/metabolism , Transcriptional Activation , 3T3-L1 Cells , Animals , CCAAT-Enhancer-Binding Proteins/genetics , Cyclin C/genetics , Humans , Mice , Mice, Knockout , PPAR gamma/genetics , PPAR gamma/metabolism
14.
Am J Physiol Renal Physiol ; 315(6): F1658-F1669, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30280599

ABSTRACT

Muscle dysfunction is an important cause of morbidity among patients with chronic kidney disease (CKD). Although muscle fibrosis is present in a CKD rodent model, its existence in humans and its impact on physical function are currently unknown. We examined isometric leg extension strength and measures of skeletal muscle fibrosis and inflammation in vastus lateralis muscle from CKD patients ( n = 10) and healthy, sedentary controls ( n = 10). Histochemistry and immunohistochemistry were used to assess muscle collagen and macrophage and fibro/adipogenic progenitor (FAP) cell populations, and RT-qPCR was used to assess muscle-specific inflammatory marker expression. Muscle collagen content was significantly greater in CKD compared with control (18.8 ± 2.1 vs. 11.7 ± 0.7% collagen area, P = 0.008), as was staining for collagen I, pro-collagen I, and a novel collagen-hybridizing peptide that binds remodeling collagen. Muscle collagen was inversely associated with leg extension strength in CKD ( r = -0.74, P = 0.01). FAP abundance was increased in CKD, was highly correlated with muscle collagen ( r = 0.84, P < 0.001), and was inversely associated with TNF-α expression ( r = -0.65, P = 0.003). TNF-α, CD68, CCL2, and CCL5 mRNA were significantly lower in CKD than control, despite higher serum TNF-α and IL-6. Immunohistochemistry confirmed fewer CD68+ and CD11b+ macrophages in CKD muscle. In conclusion, skeletal muscle collagen content is increased in humans with CKD and is associated with functional parameters. Muscle fibrosis correlated with increased FAP abundance, which may be due to insufficient macrophage-mediated TNF-α secretion. These data provide a foundation for future research elucidating the mechanisms responsible for this newly identified human muscle pathology.


Subject(s)
Isometric Contraction , Muscle Strength , Muscle Weakness/etiology , Myositis/etiology , Quadriceps Muscle/physiopathology , Renal Insufficiency, Chronic/complications , Aged , Case-Control Studies , Collagen/metabolism , Cross-Sectional Studies , Female , Fibrosis , Health Status , Humans , Inflammation Mediators/metabolism , Male , Middle Aged , Muscle Weakness/diagnosis , Muscle Weakness/metabolism , Muscle Weakness/physiopathology , Myositis/diagnosis , Myositis/metabolism , Myositis/physiopathology , Quadriceps Muscle/metabolism , Quadriceps Muscle/pathology , Renal Insufficiency, Chronic/diagnosis , Renal Insufficiency, Chronic/physiopathology , Severity of Illness Index
15.
Int J Mol Sci ; 19(1)2018 Jan 09.
Article in English | MEDLINE | ID: mdl-29315239

ABSTRACT

One of the main causes of hyperglycemia is inefficient or impaired glucose utilization by skeletal muscle, which can be exacerbated by chronic high caloric intake. Previously, we identified a natural compound, mangiferin (MGF) that improved glucose utilization in high fat diet (HFD)-induced insulin resistant mice. To further identify the molecular mechanisms of MGF action on glucose metabolism, we conducted targeted metabolomics and transcriptomics studies of glycolyic and mitochondrial bioenergetics pathways in skeletal muscle. These data revealed that MGF increased glycolytic metabolites that were further augmented as glycolysis proceeded from the early to the late steps. Consistent with an MGF-stimulation of glycolytic flux there was a concomitant increase in the expression of enzymes catalyzing glycolysis. MGF also increased important metabolites in the tricarboxylic acid (TCA) cycle, such as α-ketoglutarate and fumarate. Interestingly however, there was a reduction in succinate, a metabolite that also feeds into the electron transport chain to produce energy. MGF increased succinate clearance by enhancing the expression and activity of succinate dehydrogenase, leading to increased ATP production. At the transcriptional level, MGF induced mRNAs of mitochondrial genes and their transcriptional factors. Together, these data suggest that MGF upregulates mitochondrial oxidative capacity that likely drives the acceleration of glycolysis flux.


Subject(s)
Energy Metabolism/drug effects , Glycolysis/drug effects , Mitochondria/drug effects , Xanthones/pharmacology , Animals , Cell Line , Citric Acid Cycle/drug effects , DNA, Mitochondrial/metabolism , Diet, High-Fat , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Metabolome/drug effects , Metabolomics , Mice , Mice, Inbred C57BL , Mitochondria/genetics , Mitochondria/metabolism , Succinate Dehydrogenase/genetics , Succinate Dehydrogenase/metabolism
16.
Sheng Li Xue Bao ; 69(4): 422-428, 2017 Aug 25.
Article in Zh | MEDLINE | ID: mdl-28825100

ABSTRACT

The aim of the present study was to investigate the effects of exercise on skeletal muscle autophagy. Trains of high-frequency electrical stimulation (pulses frequency: 100 Hz) were used to stimulate sciatic nerve and consequently induce muscle contraction of the left hindlimb. The unstimulated right hindlimb muscles were taken as control. The mice were sacrificed immediately (0), 30 or 60 min after the electrical stimulation by cervical dislocation, and gastrocnemius muscles were rapidly dissected and freeze-clamped in liquid nitrogen. AMP-activated protein kinase (AMPK) and the autophagy marker protein LC3 were detected by Western blotting, and muscle atrophy related genes including atrogin-1, MuRF-1, Bnip3, Bnip3l and CathepsinL were detected by using real-time qPCR. The results showed that, at 0 min after the electrical stimulation, the activity of AMPK and LC3-II/I ratio were significantly increased in left gastrocnemius muscles, compared with those of the muscles in the right hindlimb. The levels of atrogin-1, MuRF-1, Bnip3, Bnip3l and CathepsinL mRNA expressions were up-regulated by electrical stimulation. Meanwhile, the activity of autophagy related protein, ULK1 was significantly enhanced by electrical stimulation. These results suggest that electrical stimulation of sciatic nerve may induce the skeletal muscle autophagy, and this may be regulated through AMPK/ULK1-mediated signaling pathway.


Subject(s)
Autophagy , Electric Stimulation , Muscle Contraction , Muscle, Skeletal/physiology , Physical Conditioning, Animal , Sciatic Nerve/physiology , AMP-Activated Protein Kinases/metabolism , Animals , Cathepsin L/metabolism , Male , Membrane Proteins/metabolism , Mice , Microtubule-Associated Proteins/metabolism , Mitochondrial Proteins/metabolism , Muscle Proteins/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Signal Transduction , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism
17.
J Biol Chem ; 290(41): 24772-83, 2015 Oct 09.
Article in English | MEDLINE | ID: mdl-26306048

ABSTRACT

We previously reported that the skeletal muscle-specific overexpression of Fyn in mice resulted in a severe muscle wasting phenotype despite the activation of mTORC1 signaling. To investigate the bases for the loss of muscle fiber mass, we examined the relationship between Fyn activation of mTORC1, JNK, and endoplasmic reticulum stress. Overexpression of Fyn in skeletal muscle in vivo and in HEK293T cells in culture resulted in the activation of IRE1α and JNK, leading to increased cell death. Fyn synergized with the general endoplasmic reticulum stress inducer thapsigargin, resulting in the activation of IRE1α and further accelerated cell death. Moreover, inhibition of mTORC1 with rapamycin suppressed IRE1α activation and JNK phosphorylation, resulting in protecting cells against Fyn- and thapsigargin-induced cell death. Moreover, rapamycin treatment in vivo reduced the skeletal muscle IRE1α activation in the Fyn-overexpressing transgenic mice. Together, these data demonstrate the presence of a Fyn-induced endoplasmic reticulum stress that occurred, at least in part, through the activation of mTORC1, as well as subsequent activation of the IRE1α-JNK pathway driving cell death.


Subject(s)
Endoribonucleases/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Multiprotein Complexes/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-fyn/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Death/drug effects , Endoplasmic Reticulum Stress/drug effects , HEK293 Cells , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice , Muscle, Skeletal/cytology , Proto-Oncogene Proteins c-fyn/genetics , Signal Transduction/drug effects , Thapsigargin/pharmacology
18.
Diabetes Metab Res Rev ; 31(4): 346-359, 2015 May.
Article in English | MEDLINE | ID: mdl-25613819

ABSTRACT

BACKGROUND: Infection with Trypanosoma cruzi, the protozoan parasite that causes Chagas disease, results in chronic infection that leads to cardiomyopathy with increased mortality and morbidity in endemic regions. In a companion study, our group found that a high-fat diet (HFD) protected mice from T. cruzi-induced myocardial damage and significantly reduced post-infection mortality during acute T. cruzi infection. METHODS: In the present study metabolic syndrome was induced prior to T. cruzi infection by feeding a high fat diet. Also, mice were treated with anti-diabetic drug metformin. RESULTS: In the present study, the lethality of T. cruzi (Brazil strain) infection in CD-1 mice was reduced from 55% to 20% by an 8-week pre-feeding of an HFD to induce obesity and metabolic syndrome. The addition of metformin reduced mortality to 3%. CONCLUSIONS: It is an interesting observation that both the high fat diet and the metformin, which are known to differentially attenuate host metabolism, effectively modified mortality in T. cruzi-infected mice. In humans, the metabolic syndrome, as presently construed, produces immune activation and metabolic alterations that promote complications of obesity and diseases of later life, such as myocardial infarction, stroke, diabetes, Alzheimer's disease and cancer. Using an evolutionary approach, we hypothesized that for millions of years, the channeling of host resources into immune defences starting early in life ameliorated the effects of infectious diseases, especially chronic infections, such as tuberculosis and Chagas disease. In economically developed countries in recent times, with control of the common devastating infections, epidemic obesity and lengthening of lifespan, the dwindling benefits of the immune activation in the first half of life have been overshadowed by the explosion of the syndrome's negative effects in later life.


Subject(s)
Adipose Tissue, White/immunology , Chagas Disease/immunology , Energy Metabolism/drug effects , Metabolic Syndrome/immunology , Models, Immunological , Obesity/immunology , Trypanosoma cruzi/immunology , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Adipose Tissue, White/parasitology , Adiposity/drug effects , Animals , Cell Line , Chagas Disease/blood , Chagas Disease/metabolism , Chagas Disease/parasitology , Cytokines/blood , Cytokines/metabolism , Foreskin/drug effects , Foreskin/immunology , Foreskin/metabolism , Foreskin/parasitology , Heart Ventricles/drug effects , Heart Ventricles/immunology , Heart Ventricles/metabolism , Heart Ventricles/parasitology , Humans , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Leptin/blood , Leptin/metabolism , Male , Metabolic Syndrome/drug therapy , Metabolic Syndrome/etiology , Metabolic Syndrome/parasitology , Metformin/pharmacology , Metformin/therapeutic use , Mice, Inbred Strains , Obesity/blood , Obesity/metabolism , Obesity/physiopathology , Random Allocation , Survival Analysis , Trypanosoma cruzi/drug effects , Trypanosoma cruzi/isolation & purification , Trypanosoma cruzi/pathogenicity
19.
EMBO Rep ; 14(9): 795-803, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23907538

ABSTRACT

Macroautophagy (MA) regulates cellular quality control and energy balance. For example, loss of MA in aP2-positive adipocytes converts white adipose tissue (WAT) into brown adipose tissue (BAT)-like, enhancing BAT function and thereby insulin sensitivity. However, whether MA regulates early BAT development is unknown. We report that deleting Atg7 in myogenic Myf5+ progenitors inhibits MA in Myf5-cell-derived BAT and muscle. Knock out (KO) mice have defective BAT differentiation and function. Surprisingly, their body temperature is higher due to WAT lipolysis-driven increases in fatty acid oxidation in 'Beige' cells in inguinal WAT, BAT and muscle. KO mice also present impaired muscle differentiation, reduced muscle mass and glucose intolerance. Our studies show that ATG7 in Myf5+ progenitors is required to maintain energy and glucose homeostasis through effects on BAT and muscle development. Decreased MA in myogenic progenitors with age and/or overnutrition might contribute to the metabolic defects and sarcopenia observed in these conditions.


Subject(s)
Adipose Tissue, Brown/metabolism , Autophagy , Energy Metabolism , Glucose/metabolism , Homeostasis , Muscle, Skeletal/metabolism , Myogenic Regulatory Factor 5/metabolism , Adipose Tissue, Brown/growth & development , Animals , Autophagy-Related Protein 7 , Cell Differentiation , Fatty Acids/metabolism , Mice , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Muscle, Skeletal/growth & development , Myogenic Regulatory Factor 5/genetics , Stem Cells/cytology , Stem Cells/metabolism
20.
Biochem J ; 461(2): 315-22, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24785657

ABSTRACT

Macroautophagy is a physiological cellular response to nutrient stress, which leads to the engulfment of cytosolic contents by a double-walled membrane structure, the phagophore. Phagophores seal to become autophagosomes, which then fuse with lysosomes to deliver their contents for degradation. Macroautophagy is regulated by numerous cellular factors, including the Class III PI3K (phosphoinositide 3-kinase) Vps34 (vacuolar protein sorting 34). The autophagic functions of Vps34 require its recruitment to a complex that includes Vps15, Beclin-1 and Atg14L (autophagy-related 14-like protein) and is known as Vps34 Complex I. We have now identified NRBF2 (nuclear receptor-binding factor 2) as a new member of Vps34 Complex I. NRBF2 binds to complexes that include Vps34, Vps15, Beclin-1 and ATG-14L, but not the Vps34 Complex II component UVRAG (UV radiation resistance-associated gene). NRBF2 directly interacts with Vps15 via the Vps15 WD40 domain as well as other regions of Vps15. The formation of GFP-LC3 (light chain 3) punctae and PE (phosphatidylethanolamine)-conjugated LC3 (LC3-II) in serum-starved cells was inhibited by NRBF2 knockdown in the absence and presence of lysosomal inhibitors, and p62 levels were increased. Thus NRBF2 plays a critical role in the induction of starvation-induced autophagy as a specific member of Vps34 Complex I.


Subject(s)
Autophagy/genetics , Class III Phosphatidylinositol 3-Kinases/genetics , Trans-Activators/genetics , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Proteins , Beclin-1 , Class III Phosphatidylinositol 3-Kinases/metabolism , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Humans , Lysosomes/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Phagosomes/metabolism , Protein Transport , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Trans-Activators/antagonists & inhibitors , Trans-Activators/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Vacuolar Sorting Protein VPS15/genetics , Vacuolar Sorting Protein VPS15/metabolism , Vacuoles/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL