Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Biol Rev Camb Philos Soc ; 93(1): 364-389, 2018 02.
Article in English | MEDLINE | ID: mdl-28643455

ABSTRACT

The loss of functional ß cells leads to development of diabetes. Several studies have shown that ß cells are specified through several stages of progenitors during pancreas development, each stage defined by the expression of specific transcription factors (TFs). Understanding signalling pathways that control the differentiation and specification processes during embryogenesis will facilitate efforts to obtain functional ß cells in vitro. Our current knowledge of the mechanisms involved in pancreatic ß cell development and survival under normal or diabetic conditions has come largely from animal studies. However, there are marked differences in islet structure and physiological properties between humans and animals, and not all phenotypes of human diabetes can be recapitulated in animal models. Therefore, human pluripotent stem cells (hPSCs), including human embryonic stem cells (hESCs) and human induced PSCs (hiPSCs) offer a great opportunity for increasing our understanding of the pathways regulating human pancreatic ß-cell development and survival. Furthermore, hPSCs provide a renewable source of functional pancreatic ß cells for cell replacement therapy as well as disease modelling. Herein, we discuss the signalling pathways involved in the development of pancreatic ß cells during embryogenesis. Additionally, we describe how these pathways are manipulated in vitro to differentiate hPSCs into functional ß cells. Finally, we highlight the progress that has been made for the applications of those cells in treating and modelling diabetes.


Subject(s)
Cell Differentiation/physiology , Embryonic Development/physiology , Insulin-Secreting Cells/physiology , Pluripotent Stem Cells/physiology , Animals
2.
PLoS One ; 11(7): e0154151, 2016.
Article in English | MEDLINE | ID: mdl-27441649

ABSTRACT

UNLABELLED: Membrane cholesterol levels play an important factor in regulating cell function. Sarcolemmal cholesterol is concentrated in lipid rafts and caveolae, which are flask-shaped invaginations of the plasma membrane. The scaffolding protein caveolin permits the enrichment of cholesterol in caveolae, and caveolin interactions with numerous proteins regulate their function. The purpose of this study was to determine whether acute reductions in cardiomyocyte cholesterol levels alter subcellular protein kinase activation, intracellular Ca2+ and contractility. METHODS: Ventricular myocytes, isolated from adult Sprague Dawley rats, were treated with the cholesterol reducing agent methyl-ß-cyclodextrin (MßCD, 5 mM, 1 hr, room temperature). Total cellular cholesterol levels, caveolin-3 localization, subcellular, ERK and p38 mitogen activated protein kinase (MAPK) signaling, contractility, and [Ca2+]i were assessed. RESULTS: Treatment with MßCD reduced cholesterol levels by ~45 and shifted caveolin-3 from cytoskeleton and triton-insoluble fractions to the triton-soluble fraction, and increased ERK isoform phosphorylation in cytoskeletal, cytosolic, triton-soluble and triton-insoluble membrane fractions without altering their subcellular distributions. In contrast the primary effect of MßCD was on p38 subcellular distribution of p38α with little effect on p38 phosphorylation. Cholesterol depletion increased cardiomyocyte twitch amplitude and the rates of shortening and relaxation in conjunction with increased diastolic and systolic [Ca2+]i. CONCLUSIONS: These results indicate that acute reductions in membrane cholesterol levels differentially modulate basal cardiomyocyte subcellular MAPK signaling, as well as increasing [Ca2+]i and contractility.


Subject(s)
Cholesterol/metabolism , Myocardial Contraction/physiology , Myocytes, Cardiac/metabolism , Signal Transduction , Animals , Calcium/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Intracellular Space/metabolism , Male , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/enzymology , Rats, Sprague-Dawley , Signal Transduction/drug effects , Subcellular Fractions/metabolism , beta-Cyclodextrins/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
3.
Sci Rep ; 5: 18502, 2015 Dec 21.
Article in English | MEDLINE | ID: mdl-26687026

ABSTRACT

Lipasin/Angptl8 is a feeding-induced hepatokine that regulates triglyceride (TAG) metabolism; its therapeutical potential, mechanism of action, and relation to the lipoprotein lipase (LPL), however, remain elusive. We generated five monoclonal lipasin antibodies, among which one lowered the serum TAG level when injected into mice, and the epitope was determined to be EIQVEE. Lipasin-deficient mice exhibited elevated postprandial activity of LPL in the heart and skeletal muscle, but not in white adipose tissue (WAT), suggesting that lipasin suppresses the activity of LPL specifically in cardiac and skeletal muscles. Consistently, mice injected with the effective antibody or with lipasin deficiency had increased postprandial cardiac LPL activity and lower TAG levels only in the fed state. These results suggest that lipasin acts, at least in part, in an endocrine manner. We propose the following model: feeding induces lipasin, activating the lipasin-Angptl3 pathway, which inhibits LPL in cardiac and skeletal muscles to direct circulating TAG to WAT for storage; conversely, fasting induces Angptl4, which inhibits LPL in WAT to direct circulating TAG to cardiac and skeletal muscles for oxidation. This model suggests a general mechanism by which TAG trafficking is coordinated by lipasin, Angptl3 and Angptl4 at different nutritional statuses.


Subject(s)
Angiopoietins/metabolism , Antibodies, Monoclonal/administration & dosage , Lipoprotein Lipase/biosynthesis , Peptide Hormones/immunology , Adipose Tissue/metabolism , Angiopoietin-Like Protein 3 , Angiopoietin-Like Protein 4 , Angiopoietin-Like Protein 8 , Angiopoietin-like Proteins , Animals , Antibodies, Monoclonal/immunology , Humans , Lipoprotein Lipase/immunology , Lipoprotein Lipase/metabolism , Mice , Muscle, Skeletal/metabolism , Myocardium/metabolism , Peptide Hormones/deficiency , Peptide Hormones/therapeutic use , Postprandial Period , Triglycerides/blood
4.
Cardiovasc Diabetol ; 13: 133, 2014 Sep 13.
Article in English | MEDLINE | ID: mdl-25212743

ABSTRACT

Metabolic syndrome includes glucose intolerance and dyslipidemia, both of which are strong risk factors for developing diabetes and atherosclerotic cardiovascular diseases. Recently, multiple groups independently studied a previously uncharacterized gene, officially named C19orf80 (human) and Gm6484 (mouse), but more commonly known as RIFL, Angptl8, betatrophin and lipasin. Both exciting and conflicting results have been obtained, and significant controversy is ongoing. Accumulating evidence from genome wide association studies and mouse genetic studies convincingly shows that lipasin is involved in lipid regulation. However, the mechanism of action, the identity of transcription factors mediating its nutritional regulation, circulating levels, and relationship among lipasin, Angptl3 and Angptl4, remain elusive. Betatrophin represents a promising drug target for replenishing ß-cell mass, but current results have not been conclusive regarding its potency and specificity. Here, we summarize the consensus and controversy regarding functions of lipasin/betatrophin based on currently available evidence.


Subject(s)
Glucose/metabolism , Homeostasis/physiology , Lipid Metabolism/physiology , Peptide Hormones/metabolism , Angiopoietin-Like Protein 8 , Angiopoietin-like Proteins , Animals , Genome-Wide Association Study , Humans
6.
Sci Rep ; 4: 5013, 2014 May 23.
Article in English | MEDLINE | ID: mdl-24852694

ABSTRACT

Lipasin (also known as C19ORF80, RIFL, ANGPTL8 and betatrophin) is a newly discovered circulating factor that regulates lipid metabolism and promotes pancreatic ß-cell proliferation. Whether circulating levels of lipasin in humans are altered in a) type 2 diabetes; b) obesity and c) the postprandial state, however, is unknown. The current study aimed to compare serum lipasin levels in those who were a) non-diabetic (N=15) or diabetic (BMI- and age-matched; N=14); b) lean or obese (N=53 totally) and c) fasting and 2 hours following a defined meal (N=12). Serum lipasin levels were determined by the enzyme-linked immunosorbent assay. Lipasin levels [mean±SEM] were increased by more than two fold (P<0.001) in the diabetic patients (5.56±0.73 ng/mL) as compared to the control subjects (2.19±0.24 ng/mL). Serum lipasin levels were positively correlated with BMI (rho=0.49, P<0.001), and showed a 35% increase 2 hours following a defined meal (P=0.009). Therefore, lipasin/betatrophin is nutritionally-regulated hepatokine that is increased in human type 2 diabetes and obesity.


Subject(s)
Biomarkers/blood , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/diagnosis , Obesity/blood , Obesity/diagnosis , Peptide Hormones/blood , Angiopoietin-Like Protein 8 , Angiopoietin-like Proteins , Case-Control Studies , Female , Follow-Up Studies , Humans , Male , Middle Aged , Prognosis
7.
Diabetes ; 63(6): 1933-47, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24584551

ABSTRACT

Insulin receptor substrate 1 (IRS1) is a key mediator of insulin signal transduction. Perturbations involving IRS1 complexes may lead to the development of insulin resistance and type 2 diabetes (T2D). Surprisingly little is known about the proteins that interact with IRS1 in humans under health and disease conditions. We used a proteomic approach to assess IRS1 interaction partners in skeletal muscle from lean healthy control subjects (LCs), obese insulin-resistant nondiabetic control subjects (OCs), and participants with T2D before and after insulin infusion. We identified 113 novel endogenous IRS1 interaction partners, which represents the largest IRS1 interactome in humans and provides new targets for studies of IRS1 complexes in various diseases. Furthermore, we generated the first global picture of IRS1 interaction partners in LCs, and how they differ in OCs and T2D patients. Interestingly, dozens of proteins in OCs and/or T2D patients exhibited increased associations with IRS1 compared with LCs under the basal and/or insulin-stimulated conditions, revealing multiple new dysfunctional IRS1 pathways in OCs and T2D patients. This novel abnormality, increased interaction of multiple proteins with IRS1 in obesity and T2D in humans, provides new insights into the molecular mechanism of insulin resistance and identifies new targets for T2D drug development.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 2/metabolism , Insulin Receptor Substrate Proteins/metabolism , Insulin Resistance/genetics , Muscle, Skeletal/metabolism , Obesity/metabolism , Thinness/metabolism , Body Mass Index , Diabetes Mellitus, Type 2/genetics , Female , Genetic Predisposition to Disease , Genetic Variation , Glucose Clamp Technique , Humans , Insulin/administration & dosage , Insulin Receptor Substrate Proteins/genetics , Linkage Disequilibrium , Male , Middle Aged , Obesity/genetics , Promoter Regions, Genetic , RNA, Messenger , Risk Factors , Signal Transduction , Thinness/genetics
8.
Proc Natl Acad Sci U S A ; 110(15): 5864-9, 2013 Apr 09.
Article in English | MEDLINE | ID: mdl-23533279

ABSTRACT

Agonist-induced phosphorylation of the parathyroid hormone (PTH) receptor 1 (PTHR1) regulates receptor signaling in vitro, but the role of this phosphorylation in vivo is uncertain. We investigated this role by injecting "knock-in" mice expressing a phosphorylation-deficient (PD) PTHR1 with PTH ligands and assessing acute biologic responses. Following injection with PTH (1-34), or with a unique, long-acting PTH analog, PD mice, compared with WT mice, exhibited enhanced increases in cAMP levels in the blood, as well as enhanced cAMP production and gene expression responses in bone and kidney tissue. Surprisingly, however, the hallmark hypercalcemic and hypophosphatemic responses were markedly absent in the PD mice, such that paradoxical hypocalcemic and hyperphosphatemic responses were observed, quite strikingly with the long-acting PTH analog. Spot urine analyses revealed a marked defect in the capacity of the PD mice to excrete phosphate, as well as cAMP, into the urine in response to PTH injection. This defect in renal excretion was associated with a severe, PTH-induced impairment in glomerular filtration, as assessed by the rate of FITC-inulin clearance from the blood, which, in turn, was explainable by an overly exuberant systemic hypotensive response. The overall findings demonstrate the importance in vivo of PTH-induced phosphorylation of the PTHR1 in regulating acute ligand responses, and they serve to focus attention on mechanisms that underlie the acute calcemic response to PTH and factors, such as blood phosphate levels, that influence it.


Subject(s)
Bone and Bones/metabolism , Kidney/metabolism , Parathyroid Hormone/analogs & derivatives , Receptor, Parathyroid Hormone, Type 1/physiology , Animals , Calcium/blood , Calcium/urine , Cyclic AMP/blood , Cyclic AMP/urine , Dose-Response Relationship, Drug , Gene Expression Profiling , Gene Knock-In Techniques , Homeostasis , Humans , Ligands , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphates/blood , Phosphates/urine , Phosphorylation , Rats , Receptors, G-Protein-Coupled/metabolism , Time Factors
9.
Biochem Biophys Res Commun ; 432(3): 401-5, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23415864

ABSTRACT

Patients with metabolic syndrome are at high risk for developing atherosclerotic cardiovascular diseases and diabetes. In addition to total cholesterol, LDL-C and HDL-C, elevated plasma triglycerides (TG) are increasingly recognized as an independent risk factor for cardiovascular diseases. Recently 3 groups independently reported the identification and characterization of a novel blood lipid regulator, Lipasin/RIFL/ANGPTL8, which here is referred to as Lipasin for its lipoprotein lipase inhibition effect and for being a circulating factor denoted by 'in'. Being highly enriched in the liver, Lipasin is a hepatocyte-derived circulating factor that regulates plasma TG levels. Lipasin is nutritionally regulated, as its mRNA levels in liver and fat as well as its protein level in serum are reduced by fasting. Mice deficient for Lipasin have lower serum TG levels; conversely, its adenovirus-mediated overexpression increases serum TG levels, in part, through promoting ANGPTL3 cleavage, releasing its N-terminal domain that inhibits lipoprotein lipase. Lipasin sequence variations are associated with LDL-C and HDL-C concentrations in humans. Being lipogenic, Lipasin is highly induced during adipogenesis. Levels of Lipasin and ANGPTL4 show opposite changes in response to fasting or cold environment. Lipasin, a novel but atypical ANGPTL family member, is emerging as a critical lipid regulator and a potential drug target.


Subject(s)
Adipogenesis , Lipid Metabolism , Lipoprotein Lipase/antagonists & inhibitors , Peptide Hormones/metabolism , Angiopoietin-Like Protein 3 , Angiopoietin-Like Protein 4 , Angiopoietin-Like Protein 8 , Angiopoietin-like Proteins , Angiopoietins/chemistry , Angiopoietins/metabolism , Animals , Cholesterol, HDL/blood , Cholesterol, HDL/metabolism , Cholesterol, LDL/blood , Cholesterol, LDL/metabolism , Cold Temperature , Humans , Metabolic Syndrome/complications , Metabolic Syndrome/genetics , Metabolic Syndrome/metabolism , Mice , Peptide Hormones/chemistry , Peptide Hormones/genetics , Protein Conformation , Triglycerides/blood , Triglycerides/metabolism
10.
Cell Signal ; 25(4): 981-8, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23314177

ABSTRACT

We previously have shown that Ahsg, a liver glycoprotein, inhibits insulin receptor (InsR) tyrosine kinase (TK) activity and the ERK1/2 mitogenic signaling arm of insulin signaling. Here we show that Ahsg blocks insulin-stimulated GLUT4 translocation and Akt activation in intact cells (mouse myoblasts). Furthermore, Ahsg inhibits InsR autophosphorylation of highly-purified insulin holoreceptors in a cell-free, ATP-dependent system, with an IC50 within the range of single-chain Ahsg concentrations in human serum. Binding of (125)I-insulin to living cells overexpressing the InsR shows a dissociation constant (KD) of 250pM, unaltered in the presence of 300 nM Ahsg. A mutant InsR cDNA encoding the signal peptide, the ß-subunit and the furin processing site, but deleting the α-subunit, was stably expressed in HEK293 cells. Treatment with peroxovanadate, but not insulin, dramatically increased the 95 kD ß-subunit tyrosine phosphoryation. The level of tyrosine phosphorylation of the 95-kD ß-subunit can be driven down sharply by treatment of living HEK293 transfectant cells with physiological doses of Ahsg. Treatment of myogenic cells with Ahsg blunts insulin-stimulated InsR autophosphorylation and AKT phosphorylation. Taken together, we show that Ahsg antagonizes the metabolic functions initiated by InsR activation without interference in insulin binding. The experiments suggest a direct interaction of Ahsg with the InsR ectodomain ß-subunit in a mode that does not significantly alter the high-affinity binding of insulin to the holoreceptor's two complementing α-subunits.


Subject(s)
Receptor, Insulin/metabolism , alpha-2-HS-Glycoprotein/pharmacology , Adenosine Triphosphate/metabolism , Animals , Cell Line , Cell-Free System , Glucose Transporter Type 4/metabolism , HEK293 Cells , Humans , Insulin/pharmacology , Kinetics , Mice , Mutation , Phosphorylation/drug effects , Protein Binding , Protein Subunits/genetics , Protein Subunits/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Receptor, Insulin/genetics , Signal Transduction/drug effects , Transfection
11.
Biochem Biophys Res Commun ; 430(3): 1126-31, 2013 Jan 18.
Article in English | MEDLINE | ID: mdl-23261442

ABSTRACT

Hyperlipidemia is a major contributor to cardiovascular diseases. Members of the angiopoietin-like protein family (ANGPTLs) are important determinants of blood lipid levels. Lipasin, a newly identified gene that regulates serum triglycerides, is homologous to ANGPTL3's N-terminal domain, which is sufficient and necessary for blood lipid regulation. Brown fat is critical in mediating energy homeostasis. Thermogenesis is the primary function of brown fat, in which Lipasin and some ANGPTLs are abundant; it is unknown, however, whether these genes are thermoregulated. We therefore comprehensively examined the thermoregulation of Lipasin and ANGPTLs in brown fat. Here we show that Lipasin is a novel but atypical member of the ANGPTL family because it is within the same branch as ANGPTL3 and 4 by phylogenetic analysis. The mRNA levels of Lipasin are dramatically increased in the cold environment (4 °C for 4 h) whereas those of ANGPTL4 and ANGPTL2 are suppressed. Fasting dramatically suppresses Lipasin but increases ANGPTL4. High-fat diet treatment increases Lipasin, but reduces ANGPTL2. The distinct transcriptional regulations of Lipasin, ANGPTL2 and ANGPTL4 in brown fat in response to cold exposure and nutritional stimulation suggest distinct physiological roles for ANGPTL family members in mediating thermogenesis and energy homeostasis.


Subject(s)
Adipose Tissue, Brown/physiology , Angiopoietins/metabolism , Body Temperature Regulation , Peptide Hormones/metabolism , Angiopoietin-Like Protein 8 , Angiopoietin-like Proteins , Angiopoietins/classification , Angiopoietins/genetics , Animals , Cold Temperature , Humans , Male , Mice , Mice, Inbred C57BL , Peptide Hormones/classification , Peptide Hormones/genetics , Phylogeny , Protein Structure, Tertiary
12.
J Endocrinol ; 216(3): 315-29, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23197743

ABSTRACT

Limited information is available on the role of MAPK phosphatase 1 (MKP1) signaling in osteoblasts. We have recently reported distinct roles for MKP1 during osteoblast proliferation, differentiation, and skeletal responsiveness to parathyroid hormone (PTH). As MKP1 regulates the phosphorylation status of MAPKs, we investigated the involvement of P-ERK and P-p38 MAPKs in MKP1 knockout (KO) early and mature osteoblasts with respect to mineralization and PTH response. Calvarial osteoblasts from 9-14-week-old WT and MKP1 KO male and female mice were examined. Western blot analysis revealed downregulation and sustained expressions of P-ERK and P-p38 with PTH treatment in differentiated osteoblasts derived from KO males and females respectively. Exposure of early osteoblasts to p38 inhibitor, SB203580 (S), markedly inhibited mineralization in WT and KO osteoblasts from both genders as determined by von Kossa assay. In osteoblasts from males, ERK inhibitor U0126 (U), not p38 inhibitor (S), prevented the inhibitory effects of PTH on mineralization in early or mature osteoblasts. In osteoblasts from KO females, PTH sustained mineralization in early osteoblasts and decreased mineralization in mature cells. This effect of PTH was attenuated by S in early osteoblasts and by U in mature KO cells. Changes in matrix Gla protein expression with PTH in KO osteoblasts did not correlate with mineralization, indicative of MKP1-dependent additional mechanisms essential for PTH action on osteoblast mineralization. We conclude that PTH regulation of osteoblast mineralization in female mice is maturation stage specific and involves MKP1 modulation of P-ERK and P-p38 MAPKs.


Subject(s)
Calcification, Physiologic/physiology , Dual Specificity Phosphatase 1/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Osteoblasts/metabolism , Parathyroid Hormone/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Butadienes/pharmacology , Calcification, Physiologic/drug effects , Cell Differentiation/drug effects , Down-Regulation , Dual Specificity Phosphatase 1/genetics , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Female , Imidazoles/pharmacology , Male , Mice , Mice, Knockout , Nitriles/pharmacology , Osteoblasts/drug effects , Phosphorylation/drug effects , Phosphorylation/physiology , Pyridines/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
13.
PLoS One ; 7(9): e46254, 2012.
Article in English | MEDLINE | ID: mdl-23029450

ABSTRACT

Obesity increases the risk of multiple diseases, such as type 2 diabetes and coronary heart diseases, and therefore the current obesity epidemic poses a major public health issue. Therapeutic approaches are urgently needed to treat obesity as well as its complications. Plasma-membrane proteins with restricted tissue distributions are attractive drug targets, because of their accessibility to various drug delivery mechanisms and potentially alleviated side effects. To identify genes involved in metabolism, we performed RNA-Seq on fat in mice treated with a high-fat diet or fasting. Here we show that the gene A530016L24Rik (human ortholog C14orf180), named Nrac, is a novel nutritionally-regulated adipose and cardiac-enriched gene. Nrac is expressed specifically and abundantly in fat and the heart. Both fasting and obesity reduced Nrac expression in white adipose tissue, and fasting reduced its expression in brown fat. Nrac is localized to the plasma membrane, and highly induced during adipocyte differentiation. Nrac is therefore a novel adipocyte marker and has potential functions in metabolism.


Subject(s)
Adipocytes/metabolism , Adipogenesis/genetics , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Membrane Proteins/genetics , Obesity/genetics , Adipocytes/pathology , Adipose Tissue, Brown/pathology , Adipose Tissue, White/pathology , Amino Acid Sequence , Animals , Cell Differentiation , Diet, High-Fat , Disease Models, Animal , Fasting , Gene Expression , Humans , Male , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Myocardium/metabolism , Obesity/pathology , Organ Specificity , Protein Structure, Tertiary , Sequence Homology, Amino Acid
14.
Am J Physiol Endocrinol Metab ; 302(11): E1363-72, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22414806

ABSTRACT

Previous studies have demonstrated that parathyroid hormone (PTH) binding to the PTH/PTH-related peptide receptor (PPR) stimulates G protein coupling, receptor phosphorylation, ß-arrestin translocation, and internalization of the ligand/receptor complex. The extracellular signal-regulated mitogen-activated protein kinases 1/2 (ERK1/2 MAPK) are downstream effectors of PPR. In the current study, we investigated the role of PPR phosphorylation in the PTH regulation of the ERK1/2 MAPK pathway. Short treatment with PTH (0-40 min) of LLCP-K(1) cells stably expressing a wild-type (WT) or a phosphorylation-deficient (PD) PPR (WT-PPR or PD-PPR cells, respectively) results in similar activation of ERK1/2. Interestingly, PTH stimulation of ERK1/2 in the WT-PPR cells then decreases as a result of longer PTH (60 min) treatment, and inhibition of ERK1/2 by PTH is observed at 90 min. Strikingly, the PD-PPR cells exhibit prolonged ERK1/2 activation up to 90 min of PTH treatment. An ERK1/2-dependent increase in c-fos expression is observed in the PD-PPR cells. Subsequently, c-fos expression in the WT-PPR and PD-PPR cells was markedly attenuated by a specific ERK1/2 pathway inhibitor. Further investigations revealed that PTH treatment causes a robust recruitment of a green fluorescent protein-tagged ß-arrestin2 (ß-arrestin2-GFP) in the WT-PPR cells. In contrast, ß-arrestin2 recruitment was reduced in the PD-PPR cells. Importantly, expression of a receptor phosphorylation-independent ß-arrestin2 (R169E) in the PD-PPR cells restored the biphasic effect of PTH on ERK1/2 as in the WT-PPR cells. The study reports a novel role for receptor phosphorylation and ß-arrestin2 in the subsequent inhibition of the ERK1/2 pathway and in control of gene expression.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Genes, fos/genetics , Mitogen-Activated Protein Kinases/metabolism , Parathyroid Hormone-Related Protein/physiology , Parathyroid Hormone/physiology , Animals , Arrestins/metabolism , Blotting, Western , Cell Membrane/metabolism , Cells, Cultured , Enzyme Activation , Green Fluorescent Proteins/metabolism , Inositol 1,4,5-Trisphosphate/metabolism , Kidney Tubules/cytology , Kidney Tubules/metabolism , MAP Kinase Signaling System/genetics , Microscopy, Confocal , Parathyroid Hormone/genetics , Parathyroid Hormone-Related Protein/genetics , Phosphorylation , Swine , beta-Arrestins
15.
Am J Physiol Endocrinol Metab ; 302(10): E1183-8, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22338074

ABSTRACT

Activation of G protein-coupled receptors by agonists leads to receptor phosphorylation, internalization of ligand receptor complexes, and desensitization of hormonal response. The role of parathyroid hormone (PTH) receptor 1, PTHR1, is well characterized and known to regulate cellular responsiveness in vitro. However, the role of PTHR1 phosphorylation in bone formation is yet to be investigated. We have previously demonstrated that impaired internalization and sustained cAMP stimulation of phosphorylation-deficient (PD) PTHR1 leads to exaggerated cAMP response to subcutaneous PTH infusion in a PD knockin mouse model. To understand the physiological role of receptor internalization on PTH bone anabolic action, we examined bone parameters of wild-type (WT) and PD knockin female and male mice following PTH treatment. We found a decrease in total and diaphyseal bone mineral density in female but not in male PD mice compared with WT controls at 3-6 mo of age. This effect was attenuated at older age groups. PTH administration displayed increased bone volume and trabecular thickness in the vertebrae and distal femora of both WT and PD animals. These results suggest that PTHR1 phosphorylation does not play a major role in the anabolic action of PTH.


Subject(s)
Osteogenesis/physiology , Parathyroid Hormone/metabolism , Receptor, Parathyroid Hormone, Type 1/genetics , Receptor, Parathyroid Hormone, Type 1/metabolism , Aging/physiology , Animals , Body Weight/drug effects , Body Weight/physiology , Bone Density/drug effects , Bone Density/physiology , Female , Femur/diagnostic imaging , Femur/drug effects , Femur/physiology , Gene Knock-In Techniques , Male , Mice , Mice, Mutant Strains , Osteoblasts/cytology , Osteogenesis/drug effects , Parathyroid Hormone/pharmacology , Phosphorylation/physiology , Primary Cell Culture , Skull/cytology , Skull/drug effects , Skull/physiology , X-Ray Microtomography
16.
J Endocrinol ; 212(2): 139-47, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22107955

ABSTRACT

GnRH is the main regulator of the hypothalamic-pituitary-gonadal (H-P-G) axis. GnRH stimulates the pituitary gonadotroph to synthesize and secrete gonadotrophins (LH and FSH), and this effect of GnRH is dependent on the availability of glucose and other nutrients. Little is known about whether GnRH regulates glucose metabolism in the gonadotroph. This study examined the regulation of glucose transporters (Gluts) by GnRH in the LßT2 gonadotroph cell line. Using real-time PCR analysis, the expression of Glut1, -2, -4, and -8 was detected, but Glut1 mRNA expression level was more abundant than the mRNA expression levels of Glut2, -4, and -8. After the treatment of LßT2 cells with GnRH, Glut1 mRNA expression was markedly induced, but there was no GnRH-induction of Glut2, -4, or -8 mRNA expression in LßT2 cells. The effect of GnRH on Glut1 mRNA expression is partly mediated by ERK activation. GnRH increased GLUT1 protein and stimulated GLUT1 translocation to the cell surface of LßT2 cells. Glucose uptake assays were performed in LßT2 cells and showed that GnRH stimulates glucose uptake in the gonadotroph. Finally, exogenous treatment of mice with GnRH increased the expression of Glut1 but not the expression of Glut2, -4, or -8 in the pituitary. Therefore, regulation of glucose metabolism by GnRH via changes in Gluts expression and subcellular location in the pituitary gonadotroph reveals a novel response of the gonadotroph to GnRH.


Subject(s)
Glucose Transporter Type 1/metabolism , Glucose/metabolism , Gonadotrophs/metabolism , Gonadotropin-Releasing Hormone/metabolism , Up-Regulation , Animals , Biological Transport , Cell Line , Cell Membrane/metabolism , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/chemistry , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Glucose Transport Proteins, Facilitative/genetics , Glucose Transport Proteins, Facilitative/metabolism , Glucose Transporter Type 1/genetics , Gonadotrophs/cytology , Gonadotrophs/drug effects , Male , Mice , Mice, Inbred C57BL , Phosphorylation/drug effects , Pituitary Gland/growth & development , Pituitary Gland/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , RNA, Messenger/metabolism , Up-Regulation/drug effects
17.
J Endocrinol ; 211(2): 145-56, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21852324

ABSTRACT

Parathyroid hormone (PTH) signaling via PTH 1 receptor (PTH1R) involves mitogen-activated protein kinase (MAPK) pathways. MAPK phosphatase 1 (MKP1) dephosphorylates and inactivates MAPKs in osteoblasts, the bone-forming cells. We previously showed that PTH1R activation in differentiated osteoblasts upregulates MKP1 and downregulates pERK1/2-MAPK and cyclin D1. In this study, we evaluated the skeletal phenotype of Mkp1 knockout (KO) mice and the effects of PTH in vivo and in vitro. Microcomputed tomography analysis of proximal tibiae and distal femora from 12-week-old Mkp1 KO female mice revealed osteopenic phenotype with significant reduction (8-46%) in bone parameters compared with wild-type (WT) controls. Histomorphometric analysis showed decreased trabecular bone area in KO females. Levels of serum osteocalcin (OCN) were lower and serum tartrate-resistant acid phosphatase 5b (TRAP5b) was higher in KO animals. Treatment of neonatal mice with hPTH (1-34) for 3 weeks showed attenuated anabolic responses in the distal femora of KO mice compared with WT mice. Primary osteoblasts derived from KO mice displayed delayed differentiation determined by alkaline phosphatase activity, and reduced expressions of Ocn and Runx2 genes associated with osteoblast maturation and function. Cells from KO females exhibited attenuated PTH response in mineralized nodule formation in vitro. Remarkably, this observation was correlated with decreased PTH response of matrix Gla protein expression. Expressions of pERK1/2 and cyclin D1 were inhibited dramatically by PTH in differentiated osteoblasts from WT mice but much less in osteoblasts from Mkp1 KO mice. In conclusion, MKP1 is important for bone homeostasis, osteoblast differentiation and skeletal responsiveness to PTH.


Subject(s)
Bone and Bones/metabolism , Dual Specificity Phosphatase 1/metabolism , Osteoblasts/metabolism , Parathyroid Hormone/pharmacology , Acid Phosphatase/blood , Animals , Animals, Newborn , Blotting, Western , Bone and Bones/anatomy & histology , Bone and Bones/drug effects , Cell Differentiation/drug effects , Cell Line , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Cyclin D1/genetics , Cyclin D1/metabolism , Dual Specificity Phosphatase 1/genetics , Female , Gene Expression/drug effects , Isoenzymes/blood , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Osteoblasts/cytology , Osteoblasts/drug effects , Osteocalcin/blood , Osteocalcin/genetics , Osteocalcin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tartrate-Resistant Acid Phosphatase , X-Ray Microtomography
18.
Endocr Pract ; 17(6): 867-72, 2011.
Article in English | MEDLINE | ID: mdl-21613053

ABSTRACT

OBJECTIVE: To evaluate the prevalence of low urine calcium excretion in African American patients with primary hyperparathyroidism (PHPT), a common disorder associated with bone and renal complications, and to assess the distinction between PHPT and familial hypocalciuric hypercalcemia (FHH), a rare benign genetic disease. METHODS: We conducted a retrospective study on a cohort of 1,297 patients in whom a 24-hour urine study was performed for measurement of urine calcium and creatinine. PHPT was diagnosed if the serum calcium concentration was ≥10.5 mg/dL and intact parathyroid hormone (PTH) was ≥40 pg/mL. Patients receiving medications that affect urine calcium or with glomerular filtration rate ≤30 mL/min were excluded. RESULTS: Ninety-six patients satisfied the diagnostic criteria for PHPT. The African American (n = 70) and non-African American (n = 26) patients did not differ in their mean age, body mass index, glomerular filtration rate, serum PTH, 25-hydroxyvitamin D levels, and 24-hour urine creatinine values. Median values of urine calcium/creatinine (mg/g) were 122 for African American versus 214 for non-African American patients (P = .006). Thirty-one of 70 African American patients (44%) had a urine calcium/creatinine ratio ≤100 mg/g, whereas only 2 of 26 non-African American patients (8%) had this value (P = .001). CONCLUSION: The prevalence of low urine calcium excretion among African American patients with PHPT is unexpectedly high. A threshold of 100 mg/g urine calcium/creatinine identified 44% of such patients with PHPT as having FHH in this cohort. Therefore, other clinical criteria and laboratory variables should be used to distinguish PHPT from FHH in African American patients with PTH-dependent hypercalcemia.


Subject(s)
Black or African American , Calcium/urine , Hypercalcemia/congenital , Hyperparathyroidism, Primary/urine , Aged , Algorithms , Calcium/blood , Cohort Studies , Creatinine/urine , Diagnosis, Differential , Electronic Health Records , Female , Humans , Hypercalcemia/blood , Hypercalcemia/epidemiology , Hypercalcemia/ethnology , Hypercalcemia/urine , Hyperparathyroidism, Primary/blood , Hyperparathyroidism, Primary/diagnosis , Hyperparathyroidism, Primary/ethnology , Male , Michigan/epidemiology , Middle Aged , Parathyroid Hormone/blood , Prevalence , Retrospective Studies
19.
Cell Signal ; 23(6): 980-90, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21087662

ABSTRACT

Ahsg (fetuin-A) is a 55-59kDa phosphorylated glycoprotein synthesized in the adult predominantly by hepatocytes, from which it enters the circulation. When dysregulated, this glycoprotein operates to influence the clinical sequelae of insulin resistance-type 2 diabetes and cardiovascular disease. The pathological sequelae likely arise from two separable molecular "faces" of Ahsg-one acting at the level of the insulin receptor and a second face influencing ectopic biomineralization in the intima. A detailed understanding of these two functional faces of Ahsg is not yet clear for lack of structural studies. Ahsg has a physiological role in the biomineralization of bone, which when dysregulated can lead to ectopic calcification of soft tissues in the vasculature. Ahsg has a second physiological function in regulating how insulin signals through its receptor, a transmembrane tyrosine kinase. Dysregulation of this "face" of Ahsg results in morbid sequelae such as impaired glucose disposal and fatty liver. Ahsg binds to tandem fibronectin type 3 (Fn3) domains present in the 194 amino acid residue extracellular portion of the ß-subunit of the insulin receptor, distant from the high-affinity pocket formed by two complementing α-subunits where insulin binds. Only two proteins are known to bind directly to the insulin receptor ectodomain - insulin and Ahsg - the former turns on the receptor's intrinsic tyrosine kinase (TK) activity, and the latter shuts it down. Recent X-ray crystallographic studies of the ectodomain of the insulin receptor now sharpen our understanding of the receptor's extracellular α-subunit and linked ß-subunit. Ahsg genotype and its circulating level have been correlated with body morphometrics (obese versus lean and visceral adiposity) in epidemiological studies enrolling thousands of patients. Epidemiological studies from the clinic reveal high levels of circulating Ahsg in insulin resistance and diabetes. This review endeavors to explain how one protein can mediate diverse pathologies, but specifically addresses its metabolic "face" blunting insulin receptor activity, an action leading to insulin resistance.


Subject(s)
Blood Proteins/genetics , Insulin Resistance , Insulin/metabolism , Receptor, Insulin/metabolism , Amino Acid Sequence , Animals , Blood Proteins/metabolism , Diabetes Mellitus, Type 2/metabolism , Glycoproteins/physiology , Humans , Molecular Sequence Data , Polymorphism, Single Nucleotide , Protein Binding , Protein Conformation , Receptor, Insulin/antagonists & inhibitors , Sequence Alignment , Thinness/genetics , alpha-2-HS-Glycoprotein , alpha-Fetoproteins/physiology
20.
Endocr Pract ; 17(3): 363-8, 2011.
Article in English | MEDLINE | ID: mdl-21134881

ABSTRACT

OBJECTIVE: To compare lipid profiles and glucose control in African American patients with type 2 diabetes mellitus with and without chronic hepatitis C viral (HCV) infection. METHODS: This retrospective study conducted in an academic outpatient setting included African American patients with both type 2 diabetes and HCV, patients with HVC only, and patients with type 2 diabetes only. Serum total cholesterol, low-density lipoprotein (LDL) cholesterol, high-density lipoprotein (HDL) cholesterol, and triglyceride values were compared among all 3 patient groups. RESULTS: The study population included 283 patients, of whom 111 had type 2 diabetes and HCV, 68 had HCV only, and 104 had type 2 diabetes only. Chronic HCV was associated with lower total cholesterol and LDL-cholesterol levels in patients with or without type 2 diabetes. In contrast, elevated serum triglyceride levels associated with diabetes were not reduced in patients with chronic HCV, although diabetes control was better in the diabetes group with HCV than in the diabetes group without HCV (mean hemoglobin A1c [standard error of the mean]: 7.1% [1.8%]vs 8.8% [2.1%], P<.001). HDL cholesterol was higher in the patients with earlier stages of HCV when compared with HDL cholesterol in the other 2 groups. CONCLUSIONS: Chronic HCV infection in type 2 diabetic patients decreases serum levels of total and LDL cholesterol, but has no such protective effect on triglyceride levels. HCV infection may alter the cellular pathways of cholesterol and triglyceride metabolism in patients with type 2 diabetes.


Subject(s)
Black or African American , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Hepatitis C, Chronic/blood , Hepatitis C, Chronic/metabolism , Lipid Metabolism , Adolescent , Adult , Aged , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/ethnology , Female , Hepacivirus/physiology , Hepatitis C, Chronic/complications , Hepatitis C, Chronic/ethnology , Humans , Lipid Metabolism/physiology , Male , Middle Aged , Retrospective Studies , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...