Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Nutrients ; 13(10)2021 Oct 03.
Article in English | MEDLINE | ID: mdl-34684500

ABSTRACT

There is a paucity of data on whether Australian university students are meeting specific nutrient guidelines, and the relationship between diet and physical activity patterns with body composition and metabolic health. In this study, biomedical students from The University of Queensland were recruited (150 males and 211 females, 19-25 years), and nutritional intake (ASA24-Australia) and physical activity levels (Active Australia Survey) quantified. Body composition (height, waist circumference, body mass, BMI, and percentage body fat; BOD POD) and metabolic health (oral glucose tolerance test) were also measured. Median daily energy intake was 6760 kJ in females and 10,338 kJ in males, with more than 30% of total energy coming from energy-dense, nutrient-poor foods. Only 1 in 10 students met fruit or vegetable recommendations, with less than one third meeting recommendations for fibre, calcium, and potassium. Intakes of calcium and iron were particularly low among female students, with only 16% and 6% of students meeting the recommended dietary intake (RDI), respectively. The majority of males and almost half of all females exceeded the suggested dietary target (SDT) for sodium. Sufficient physical activity (≥150 min over ≥5 sessions per week) was met by more than 80% of students. Body composition and blood glucose concentrations were largely normal but an early sign of insulin resistance (HOMA-IR > 2.0), measured in a subset of students, was present in 21% of males and 17% of females. Modest reductions in blood glucose levels and percentage body fat were associated with increasing vigorous activity. Low intakes of fibre, calcium, and potassium could be corrected by increasing fruit, vegetable, and dairy intake, and, among females, health promotion messages focusing on iron-rich foods should be prioritised. While these nutrient deficiencies did not translate into immediate metabolic heath concerns, dietary behaviours can track into adulthood and have lasting effects on overall health.


Subject(s)
Body Composition , Diet, Healthy/statistics & numerical data , Exercise , Guideline Adherence/statistics & numerical data , Students, Medical/statistics & numerical data , Adult , Australia , Blood Glucose/analysis , Body Mass Index , Cross-Sectional Studies , Diet, Healthy/standards , Eating , Feeding Behavior , Female , Glucose Tolerance Test , Humans , Male , Nutrition Policy , Waist Circumference
2.
Proteomics Clin Appl ; 15(2-3): e2000078, 2021 05.
Article in English | MEDLINE | ID: mdl-33641263

ABSTRACT

PURPOSE: To identify glucocorticoid-responsive proteins measurable in human serum that may have clinical utility in therapeutic drug monitoring and the diagnosis of cortisol excess or deficiency. EXPERIMENTAL DESIGN: A phased biomarker discovery strategy was conducted in two cohorts. Secretome from peripheral blood mononuclear cells (PBMC) isolated from six volunteers after ex vivo incubation ± dexamethasone (DEX) 100 ng/mL for 4 h and 24 h was used for candidate discovery and qualification using untargeted proteomics and a custom multiple reaction monitoring mass spectrometry (MRM-MS) assay, respectively. For validation, five candidates were measured by immunoassay in serum from an independent cohort (n = 20), sampled at 1200 h before and after 4 mg oral DEX. RESULTS: The discovery secretome proteomics data generated a shortlist of 45 candidates, with 43 measured in the final MRM-MS assay. Differential analysis revealed 16 proteins that were significant in at least one of two time points. In the validation cohort, 3/5 serum proteins were DEX-responsive, two significantly decreased: lysozyme C (p < 0.0001) and nucleophosmin-1 (p < 0.01), while high mobility group box 2 significantly increased (p < 0.01). CONCLUSIONS AND CLINICAL RELEVANCE: Using an ex vivo proteomic approach in PBMC, we have identified circulating glucocorticoid-responsive proteins which may have potential as serum biomarkers of glucocorticoid activity.


Subject(s)
Glucocorticoids
3.
Biomolecules ; 10(5)2020 05 16.
Article in English | MEDLINE | ID: mdl-32429496

ABSTRACT

Esophageal adenocarcinoma (EAC) incidence has been rapidly increasing, potentially associated with the prevalence of the risk factors gastroesophageal reflux disease (GERD), obesity, high-fat diet (HFD), and the precursor condition Barrett's esophagus (BE). EAC development occurs over several years, with stepwise changes of the squamous esophageal epithelium, through cardiac metaplasia, to BE, and then EAC. To establish the roles of GERD and HFD in initiating BE, we developed a dietary intervention model in C57/BL6 mice using experimental HFD and GERD (0.2% deoxycholic acid, DCA, in drinking water), and then analyzed the gastroesophageal junction tissue lipidome and microbiome to reveal potential mechanisms. Chronic (9 months) HFD alone induced esophageal inflammation and metaplasia, the first steps in BE/EAC pathogenesis. While 0.2% deoxycholic acid (DCA) alone had no effect on esophageal morphology, it synergized with HFD to increase inflammation severity and metaplasia length, potentially via increased microbiome diversity. Furthermore, we identify a tissue lipid signature for inflammation and metaplasia, which is characterized by elevated very-long-chain ceramides and reduced lysophospholipids. In summary, we report a non-transgenic mouse model, and a tissue lipid signature for early BE. Validation of the lipid signature in human patient cohorts could pave the way for specific dietary strategies to reduce the risk of BE in high-risk individuals.


Subject(s)
Adenocarcinoma/etiology , Barrett Esophagus/etiology , Diet, High-Fat/adverse effects , Disease Models, Animal , Esophageal Neoplasms/etiology , Lipid Metabolism , Adenocarcinoma/metabolism , Animals , Barrett Esophagus/metabolism , Barrett Esophagus/pathology , Deoxycholic Acid/toxicity , Esophageal Mucosa/metabolism , Esophageal Mucosa/pathology , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Gastrointestinal Microbiome , Male , Mice , Mice, Inbred C57BL
4.
Clin Endocrinol (Oxf) ; 91(6): 728-736, 2019 12.
Article in English | MEDLINE | ID: mdl-31612515

ABSTRACT

OBJECTIVE: Thrombospondin-1 (TSP1), a matricellular protein, and Osteocalcin (OCN), a noncollagenous protein secreted by osteoblasts, are known to be up- and down-regulated, respectively, by glucocorticoids. The aim of this study was to determine whether a ratio between TSP1:OCN was altered by changes in glucocorticoid activity in humans. DESIGN: Prospective observational study. SETTING: Tertiary university hospital in Queensland, Australia. PATIENTS AND MEASUREMENTS: Patients with Cushing's syndrome (CS, n = 19), asthma or giant cell arteritis on chronic prednisolone treatment (PRED, n = 13), adrenal insufficiency (AI, n = 16) and healthy volunteers (HV, n = 20). Plasma TSP1 and serum total OCN were measured by immunoassay at 0800h, 1200h and 1600h in patients with CS, patients with AI taking replacement glucocorticoids, HV before and after 4 mg dexamethasone and PRED patients predose at 800 and 4 hours post-dose at 1200 hours. RESULTS: Plasma TSP1 in CS was higher (P < .0001), and serum OCN was lower (P < .0001) than HV. The TSP1:OCN ratio in HV increased significantly after 4 mg dexamethasone (P < .0001) and in AI after taking their hydrocortisone replacement therapy (P < .001). PRED patients had a higher TSP1:OCN ratio compared with HV at both 800 and 1200 hours (both P < .001), but no significant change occurred from pre- to post-dose. A TSP1:OCN ratio of >73 at 800 hours differentiated CS from HV with a sensitivity of 95% and a specificity of 100%. CONCLUSIONS: The TSP1:OCN ratio is elevated in patients on prednisolone and in patients with CS compared with healthy volunteers. It may be a useful biomarker of total body glucocorticoid activity in humans.


Subject(s)
Glucocorticoids/therapeutic use , Osteocalcin/blood , Thrombospondin 1/blood , Adrenal Insufficiency/blood , Adrenal Insufficiency/drug therapy , Adult , Aged , Asthma/blood , Asthma/drug therapy , Cushing Syndrome/blood , Cushing Syndrome/drug therapy , Female , Healthy Volunteers , Humans , Hydrocortisone/blood , Male , Middle Aged , Prednisolone/therapeutic use , Prospective Studies , Treatment Outcome , Young Adult
5.
BMJ Open ; 9(4): e025568, 2019 04 20.
Article in English | MEDLINE | ID: mdl-31005923

ABSTRACT

INTRODUCTION: Inflammatory bowel disease (IBD) is a chronic autoinflammatory disease of the gastrointestinal tract with peak age of onset during adolescence and young adulthood. Adolescents and young adults (AYAs) with IBD experience higher depression rates compared with peers who are well or have other chronic conditions. Mindfulness-based interventions are of particular interest because of their potential to improve both the course of IBD and depression. METHODS AND ANALYSIS: This study is a parallel design, single-blind, pilot randomised controlled trial (RCT) of mindfulness-based cognitive therapy (MBCT) in AYAs with IBD and depression. The trial aims to recruit 64 participants who will be randomly allocated to MBCT or treatment as usual. The primary outcome measure is the depression subscale score from the Depression, Anxiety and Stress Scale. Secondary outcomes include anxiety, stress, post-traumatic growth, IBD-related quality of life, illness knowledge, medication adherence, mindfulness, IBD activity, inflammatory markers, microbiome and brain neuroconnectivity changes. All outcomes other than neuroimaging will be collected at three time points: at baseline, at therapy completion and at 20 weeks. Neuroimaging will be conducted at baseline and at therapy completion. Mixed-effects linear and logistic regression modelling will be used to analyse continuous and dichotomous outcomes, respectively. Participants' experiences will be explored through focus groups, and thematic analysis will be used to generate relevant themes. ETHICS AND DISSEMINATION: The protocol has been approved by the Mater Hospital Human Research Ethics Committee (HREC) and University of Queensland HREC. Trial findings will be published in peer-reviewed journals and will be presented at scientific conferences. TRIAL REGISTRATION NUMBER: ACTRN12617000876392, U1111-1197-7370; Pre-results.


Subject(s)
Depression/therapy , Inflammatory Bowel Diseases/therapy , Mindfulness/methods , Adolescent , Adult , Depression/complications , Female , Humans , Inflammatory Bowel Diseases/complications , Male , Pilot Projects , Randomized Controlled Trials as Topic , Single-Blind Method , Young Adult
6.
FASEB J ; 33(5): 6412-6430, 2019 05.
Article in English | MEDLINE | ID: mdl-30779881

ABSTRACT

Growth hormone (GH) has an important function as an insulin antagonist with elevated insulin sensitivity evident in humans and mice lacking a functional GH receptor (GHR). We sought the molecular basis for this sensitivity by utilizing a panel of mice possessing specific deletions of GHR signaling pathways. Metabolic clamps and glucose homeostasis tests were undertaken in these obese adult C57BL/6 male mice, which indicated impaired hepatic gluconeogenesis. Insulin sensitivity and glucose disappearance rate were enhanced in muscle and adipose of mice lacking the ability to activate the signal transducer and activator of transcription (STAT)5 via the GHR (Ghr-391-/-) as for GHR-null (GHR-/-) mice. These changes were associated with a striking inhibition of hepatic glucose output associated with altered glycogen metabolism and elevated hepatic glycogen content during unfed state. The enhanced hepatic insulin sensitivity was associated with increased insulin receptor ß and insulin receptor substrate 1 activation along with activated downstream protein kinase B signaling cascades. Although phosphoenolpyruvate carboxykinase (Pck)-1 expression was unchanged, its inhibitory acetylation was elevated because of decreased sirtuin-2 expression, thereby promoting loss of PCK1. Loss of STAT5 signaling to defined chromatin immunoprecipitation targets would further increase lipogenesis, supporting hepatosteatosis while lowering glucose output. Finally, up-regulation of IL-15 expression in muscle, with increased secretion of adiponectin and fibroblast growth factor 1 from adipose tissue, is expected to promote insulin sensitivity.-Chhabra, Y., Nelson, C. N., Plescher, M., Barclay, J. L., Smith, A. G., Andrikopoulos, S., Mangiafico, S., Waxman, D. J., Brooks, A. J., Waters, M. J. Loss of growth hormone-mediated signal transducer and activator of transcription 5 (STAT5) signaling in mice results in insulin sensitivity with obesity.


Subject(s)
Carrier Proteins , Fatty Liver , Insulin Resistance/genetics , Liver , Obesity , STAT5 Transcription Factor/deficiency , Signal Transduction/genetics , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Fatty Liver/genetics , Fatty Liver/metabolism , Fatty Liver/pathology , Glucose/genetics , Glucose/metabolism , Glycogen/genetics , Glycogen/metabolism , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Knockout , Obesity/genetics , Obesity/metabolism , Obesity/pathology , Phosphoenolpyruvate Carboxykinase (GTP)/genetics , Phosphoenolpyruvate Carboxykinase (GTP)/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , STAT5 Transcription Factor/metabolism
7.
Endocr Rev ; 40(1): 66-95, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30169559

ABSTRACT

A plethora of physiological processes show stable and synchronized daily oscillations that are either driven or modulated by biological clocks. A circadian pacemaker located in the suprachiasmatic nucleus of the ventral hypothalamus coordinates 24-hour oscillations of central and peripheral physiology with the environment. The circadian clockwork involved in driving rhythmic physiology is composed of various clock genes that are interlocked via a complex feedback loop to generate precise yet plastic oscillations of ∼24 hours. This review focuses on the specific role of the core clockwork gene Period1 and its paralogs on intra-oscillator and extra-oscillator functions, including, but not limited to, hippocampus-dependent processes, cardiovascular function, appetite control, as well as glucose and lipid homeostasis. Alterations in Period gene function have been implicated in a wide range of physical and mental disorders. At the same time, a variety of conditions including metabolic disorders also impact clock gene expression, resulting in circadian disruptions, which in turn often exacerbates the disease state.


Subject(s)
Circadian Clocks/physiology , Circadian Rhythm/physiology , Homeostasis/physiology , Period Circadian Proteins/physiology , Signal Transduction/physiology , Animals , Circadian Clocks/genetics , Circadian Rhythm/genetics , Homeostasis/genetics , Humans , Period Circadian Proteins/genetics , Signal Transduction/genetics
8.
Mol Metab ; 6(6): 512-523, 2017 06.
Article in English | MEDLINE | ID: mdl-28580282

ABSTRACT

OBJECTIVE: Circadian Clock gene mutant mice show dampened 24-h feeding rhythms and an increased sensitivity to high-fat diet (HFD) feeding. Restricting HFD access to the dark phase counteracts its obesogenic effect in wild-type mice. The extent to which altered feeding rhythms are causative for the obesogenic phenotype of Clock mutant mice, however, remains unknown. METHODS: Metabolic parameters of wild-type (WT) and ClockΔ19 mutant mice (MT) were investigated under ad libitum and nighttime restricted HFD feeding. Liver circadian clock function was partially rescued by hydrodynamic tail vein delivery of WT-Clock DNA vectors in mutant mice and transcriptional, metabolic, endocrine and behavioral rhythms studied. RESULTS: Nighttime-restricted feeding restored food intake, but not body weight regulation in MT mice under HFD, suggesting Clock-dependent metabolic dysregulation downstream of circadian appetite control. Liver-directed Clock gene therapy partially restored liver circadian oscillator function and transcriptome regulation without affecting centrally controlled circadian behaviors. Under HFD, MT mice with partially restored liver clock function (MT-LR) showed normalized body weight gain, rescued 24-h food intake rhythms, and WT-like energy expenditure. This was associated with decreased nighttime leptin and daytime ghrelin levels, reduced hepatic lipid accumulation, and improved glucose tolerance. Transcriptome analysis revealed that hepatic Clock rescue in MT mice affected a range of metabolic pathways. CONCLUSION: Liver Clock gene therapy improves resistance against HFD-induced metabolic impairments in mice with circadian clock disruption. Restoring or stabilizing liver clock function might be a promising target for therapeutic interventions in obesity and metabolic disorders.


Subject(s)
CLOCK Proteins/genetics , Diet, High-Fat/adverse effects , Genetic Therapy , Hyperphagia/therapy , Liver/metabolism , Obesity/prevention & control , Animals , CLOCK Proteins/metabolism , Feeding Behavior , Hyperphagia/complications , Male , Mice , Mice, Inbred C57BL , Mutation , Obesity/etiology
9.
Sleep ; 40(6)2017 06 01.
Article in English | MEDLINE | ID: mdl-28444394

ABSTRACT

Study objectives: Shortened or mistimed sleep affects metabolic homeostasis, which may in part be mediated by dysregulation of endogenous circadian clocks. In this study, we assessed the contribution of sleep disruption to metabolic dysregulation by analysing diurnal transcriptome regulation in metabolic tissues of mice subjected to a sleep restriction (SR) paradigm. Methods: Male mice were subjected to 2 × 5 days of SR with enforced waking during the first 6 hours of the light phase. SR and control mice were sacrificed at different time points of the day and RNA preparations from the mediobasal hypothalamus (MBH), liver, and epididymal white adipose tissue (eWAT) were subjected to whole-genome microarray hybridization. Transcriptional rhythms were associated with changes in behavioral and physiological parameters such as sleep, body temperature, and food intake. Rhythm detection was performed with CircWave and transcription profiles were compared by 2-way analysis of variance and t-tests with Benjamini-Hochberg corrections. Results: Clock gene rhythms were blunted in all tissues, while transcriptome regulation was associated with either clock gene expression, sleep patterns, or food intake in a tissue-specific manner. Clock gene expression was associated with apoptosis pathways in the MBH and with tumor necrosis factor alpha signalling in liver. Food intake-associated genes included cilium movement genes in the MBH and lipid metabolism-associated transcripts in liver. Conclusions: In mice, repeated SR profoundly alters behavioral and molecular diurnal rhythms, disrupting essential signalling pathways in MBH, liver, and eWAT, which may underlie the metabolic and cognitive disturbances observed in sleep-restricted humans such as shift workers.


Subject(s)
Circadian Rhythm/genetics , Organ Specificity/genetics , Sleep Deprivation/genetics , Transcriptome , Adipose Tissue, White/metabolism , Animals , Apoptosis/genetics , Body Temperature/genetics , Circadian Clocks/genetics , Eating/genetics , Gene Expression Profiling , Gene Expression Regulation , Hypothalamus/metabolism , Lipid Metabolism/genetics , Liver/metabolism , Male , Mice , Sleep/genetics , Tumor Necrosis Factor-alpha/metabolism
10.
FASEB J ; 30(7): 2528-40, 2016 07.
Article in English | MEDLINE | ID: mdl-27006448

ABSTRACT

Adipose tissue expansion occurs through a combination of hypertrophy of existing adipocytes and generation of new adipocytes via the process of hyperplasia, which involves the proliferation and subsequent differentiation of preadipocytes. Deficiencies in hyperplasia contribute to adipose tissue dysfunction and the association of obesity with chronic cardiometabolic diseases. Thus, increased understanding of hyperplastic pathways may be expected to afford novel therapeutic strategies. We have reported that fibroblast growth factor (FGF)-1 promotes proliferation and differentiation of human preadipocytes and recently demonstrated that bone morphogenetic protein and activin membrane-bound inhibitor (BAMBI) is a central, proximal effector. Herein, we describe the identification and characterization of carboxypeptidase X (CPX)-1, a secreted collagen-binding glycoprotein, as a novel downstream effector in human primary and Simpson-Golabi-Behmel syndrome preadipocytes. CPX-1 expression increased after treatment of preadipocytes with FGF-1, BAMBI knockdown, or induction of differentiation. CPX-1 knockdown compromised preadipocyte differentiation coincident with reduced collagen expression. Furthermore, preadipocytes differentiated on matrix derived from CPX-1 knockdown cells exhibited reduced Glut4 expression and insulin-stimulated glucose uptake. Finally, CPX-1 expression was increased in adipose tissue from obese mice and humans. Collectively, these findings establish CPX-1 as a positive regulator of adipogenesis situated downstream of FGF-1/BAMBI that may contribute to hyperplastic adipose tissue expansion via affecting extracellular matrix remodeling.-Kim, Y.-H., Barclay, J. L., He, J., Luo, X., O'Neill, H. M., Keshvari, S., Webster, J. A., Ng, C., Hutley, L. J., Prins, J. B., Whitehead, J. P. Identification of carboxypeptidase X (CPX)-1 as a positive regulator of adipogenesis.


Subject(s)
Adipogenesis/physiology , Adipose Tissue/metabolism , Gene Expression Regulation/physiology , Glycoproteins/metabolism , Metalloexopeptidases/metabolism , Adipocytes/metabolism , Adipocytes/physiology , Adipogenesis/drug effects , Adult , Animals , Cell Differentiation , Dietary Fats/administration & dosage , Dietary Fats/adverse effects , Female , Fibroblast Growth Factor 1/genetics , Fibroblast Growth Factor 1/metabolism , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Glycoproteins/genetics , Humans , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Metalloexopeptidases/genetics , Mice , Middle Aged , Obesity/etiology , Obesity/metabolism
11.
Biochem Biophys Res Commun ; 473(1): 47-53, 2016 Apr 22.
Article in English | MEDLINE | ID: mdl-26995091

ABSTRACT

Apoptosis repressor with caspase recruitment domain (ARC), an endogenous inhibitor of apoptosis, is upregulated in a number of human cancers, thereby conferring drug resistance and giving a rationale for the inhibition of ARC to overcome drug resistance. Our hypothesis was that ARC would be similarly upregulated and targetable for therapy in renal cell carcinoma (RCC). Expression of ARC was assessed in 85 human RCC samples and paired non-neoplastic kidney by qPCR and immunohistochemistry, as well as in four RCC cell lines by qPCR, Western immunoblot and confocal microscopy. Contrary to expectations, ARC was significantly decreased in the majority of clear cell RCC and in three (ACHN, Caki-1 and 786-0) of the four RCC cell lines compared with the HK-2 non-cancerous human proximal tubular epithelial cell line. Inhibition of ARC with shRNA in the RCC cell line (SN12K1) that had shown increased ARC expression conferred resistance to Sunitinib, and upregulated interleukin-6 (IL-6) and vascular endothelial growth factor (VEGF). We therefore propose that decreased ARC, particularly in clear cell RCC, confers resistance to targeted therapy through restoration of tyrosine kinase-independent alternate angiogenesis pathways. Although the results are contrary to expectations from other cancer studies, they were confirmed here with multiple analytical methods. We believe the highly heterogeneous nature of cancers like RCC predicate that expression patterns of molecules must be interpreted in relation to respective matched non-neoplastic regions. In the current study, this procedure indicated that ARC is decreased in RCC.


Subject(s)
Carcinoma, Renal Cell/metabolism , Cytoskeletal Proteins/metabolism , Drug Resistance, Neoplasm , Indoles/therapeutic use , Kidney Neoplasms/metabolism , Neovascularization, Pathologic , Nerve Tissue Proteins/metabolism , Pyrroles/therapeutic use , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/chemistry , Apoptosis , Cell Line, Tumor , Cell Survival , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Immunoblotting , Immunohistochemistry , Male , Microscopy, Confocal , Middle Aged , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Sunitinib , Vascular Endothelial Growth Factor A/metabolism
12.
Ann Clin Biochem ; 53(Pt 5): 606-10, 2016 Sep.
Article in English | MEDLINE | ID: mdl-26748102

ABSTRACT

BACKGROUND: Thrombospondin-1 (TSP-1) is a circulating matricellular glycoprotein produced from many cell types including platelets. Currently TSP-1 is measured in either plasma or serum, using expensive commercial assays. AIM: To develop and validate a cost effective in-house immunoassay for human TSP-1 suitable for quantitating levels from both plasma and serum. METHODS: An in-house enzyme-linked immunosorbent assay (ELISA) was developed for the measurement of human TSP-1. Sixteen healthy volunteers (8 male and 8 female), mean age 29 years (range 21-49), body mass index (BMI) mean 23.3 kg/m(2) (range 17.3-26.7) had non-fasted venous blood sampled at 0800 h and 1600 h for both plasma and serum TSP-1. RESULTS: The assay limit of quantitation was 7.8 µg/L, inter assay CV was 17-31%, intra assay CV was 3-4% for plasma and <9% for serum. Plasma TSP-1 ranged from 133 to 478 µg/L (mean concentration 290 µg/L) in normal volunteers. Serum TSP-1 was approximately 100-fold higher, ranging from 13,700 to 44,400 µg/L (mean concentration 257,00 µg/L). There was no correlation between plasma and serum TSP-1. CONCLUSIONS: TSP-1 can be readily measured in human plasma using ELISA. Serum concentrations are 100-fold higher, reflecting documented TSP-1 release by platelets, and does not provide a meaningful measure of circulating concentrations.


Subject(s)
Thrombospondin 1/blood , Adult , Enzyme-Linked Immunosorbent Assay , Female , Humans , Limit of Detection , Male , Middle Aged , Young Adult
13.
Eur J Endocrinol ; 174(2): 193-201, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26578641

ABSTRACT

OBJECTIVE: Thrombospondin-1 (TSP1) is a matricellular protein whose gene expression has previously been shown to increase acutely after exposure to dexamethasone in vitro. The aim of this study was to determine if TSP1 is altered by acute and chronic states of glucocorticoid excess in human subjects. DESIGN AND METHODS: Three studies have been undertaken to assess the difference or change in TSP1 in response to altered glucocorticoid activity: i) an acute interventional study assessed the effects of a single 4 mg dose of dexamethasone in 20 healthy volunteers; ii) a cross-sectional study compared plasma TSP1 in 20 healthy volunteers and eight patients with Cushing's syndrome; iii) an interventional study assessed the effect on plasma TSP1 of an increase in hydrocortisone dose from ≤20 mg/day to 30 mg/day for 7 days in 16 patients with secondary adrenal insufficiency. RESULTS: In healthy volunteers, 4 mg dexamethasone significantly increased peripheral blood mononuclear cell (PBMC) TSP1 mRNA levels (P<0.0001) and plasma TSP1 concentrations (P<0.0001), peaking at 12 h. Median (interquartile range) plasma TSP1 was higher in Cushing's, 638 (535-756) ng/ml, than in healthy volunteers, 272 (237-336) ng/ml (P<0.0001). Plasma TSP1 >400 ng/ml diagnosed Cushing's syndrome with sensitivity of 100% and specificity of 85%. The higher hydrocortisone dose increased plasma TSP1 from 139 (86-199) to 256 (133-516) ng/ml, (P<0.01) in patients with secondary adrenal insufficiency. CONCLUSIONS: TSP1 is a glucocorticoid responsive protein in humans. Further research is required to determine if plasma TSP1 has a role as a glucocorticoid biomarker.


Subject(s)
Adrenal Insufficiency/blood , Cushing Syndrome/blood , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Hydrocortisone/pharmacology , Thrombospondin 1/blood , Thrombospondin 1/drug effects , Adrenal Insufficiency/drug therapy , Adult , Aged , Aged, 80 and over , Biomarkers/blood , Cross-Sectional Studies , Dexamethasone/administration & dosage , Female , Glucocorticoids/administration & dosage , Humans , Hydrocortisone/administration & dosage , Male , Middle Aged , Sensitivity and Specificity , Young Adult
14.
Growth Factors ; 34(5-6): 210-216, 2016 12.
Article in English | MEDLINE | ID: mdl-28209092

ABSTRACT

Fibroblast growth factor-1 (FGF-1) promotes differentiation of human preadipocytes into mature adipocytes via modulation of a BMP and Activin Membrane-Bound Inhibitor (BAMBI)/Peroxisome proliferator-activated receptor (PPARγ)-dependent network. Here, we combined transcriptomic and functional investigations to identify novel downstream effectors aligned with complementary analyses of gene expression in human adipose tissue to explore relationships with insulin sensitivity. RNA-Seq and qRT-PCR analysis revealed significant down-regulation of carboxypeptidase A4 (CPA4) following FGF-1 treatment or induction of differentiation of human preadipocytes in a BAMBI/PPARγ-independent manner. siRNA-mediated knockdown of CPA4 resulted in enhanced differentiation of human preadipocytes. Furthermore, expression of CPA4 in subcutaneous adipose tissue correlated negatively with indices of local and systemic (liver and muscle) insulin sensitivity. These results identify CPA4 as a negative regulator of adipogenesis that is down-regulated by FGF-1 and a putative deleterious modulator of local and systemic insulin sensitivity. Further investigations are required to define the molecular mechanism(s) involved and potential therapeutic opportunities.


Subject(s)
Adipocytes/metabolism , Adipogenesis , Carboxypeptidases A/metabolism , Fibroblast Growth Factor 1/pharmacology , Insulin Resistance , Adipocytes/cytology , Adipocytes/drug effects , Adult , Carboxypeptidases A/genetics , Cells, Cultured , Down-Regulation , Humans , Liver/metabolism , Male , Membrane Proteins/metabolism , Middle Aged , Muscle, Skeletal/metabolism , PPAR gamma/metabolism
15.
Mol Cell Endocrinol ; 413: 209-16, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26143632

ABSTRACT

Adiponectin is a salutary adipokine and hypoadiponectinemia is implicated in the aetiology of obesity-related inflammation and cardiometabolic disease making therapeutic strategies to increase adiponectin attractive. Emerging evidence, predominantly from preclinical studies, suggests induction of heme-oxygenase-1 (HO-1) increases adiponectin production and reduces inflammatory tone. Here, we aimed to test whether induction of HO-1 enhanced adiponectin production from mature adipocytes. Treatment of human adipocytes with cobalt protoporphyrin (CoPP) or hemin for 24-48 h increased HO-1 expression and activity without affecting adiponectin expression and secretion. Treatment of adipocytes with TNFα reduced adiponectin secretion and increased expression and secretion of additional pro-inflammatory cytokines, IL-6 and MCP-1, as well as expression of sXBP-1, a marker of ER stress. HO-1 induction failed to reverse these effects. These results demonstrate that induction of HO-1 does not directly enhance adiponectin production or ameliorate the pro-inflammatory effects of TNFα and argue against a direct HO-1 - adiponectin axis.


Subject(s)
Adipocytes/metabolism , Adiponectin/biosynthesis , Heme Oxygenase-1/biosynthesis , Adipocytes/cytology , Cells, Cultured , Chemokine CCL2/metabolism , Enzyme Induction/drug effects , Enzyme Induction/physiology , Humans , Interleukin-6/metabolism , Protoporphyrins/pharmacology , Tumor Necrosis Factor-alpha/metabolism
16.
Elife ; 4: e06253, 2015 Mar 30.
Article in English | MEDLINE | ID: mdl-25821984

ABSTRACT

Circadian clocks coordinate 24-hr rhythms of behavior and physiology. In mammals, a master clock residing in the suprachiasmatic nucleus (SCN) is reset by the light-dark cycle, while timed food intake is a potent synchronizer of peripheral clocks such as the liver. Alterations in food intake rhythms can uncouple peripheral clocks from the SCN, resulting in internal desynchrony, which promotes obesity and metabolic disorders. Pancreas-derived hormones such as insulin and glucagon have been implicated in signaling mealtime to peripheral clocks. In this study, we identify a novel, more direct pathway of food-driven liver clock resetting involving oxyntomodulin (OXM). In mice, food intake stimulates OXM secretion from the gut, which resets liver transcription rhythms via induction of the core clock genes Per1 and 2. Inhibition of OXM signaling blocks food-mediated resetting of hepatocyte clocks. These data reveal a direct link between gastric filling with food and circadian rhythm phasing in metabolic tissues.


Subject(s)
Circadian Clocks/drug effects , Circadian Rhythm/drug effects , Liver/drug effects , Oxyntomodulin/pharmacology , Period Circadian Proteins/genetics , Animals , Circadian Clocks/genetics , Circadian Rhythm/genetics , Eating/drug effects , Eating/physiology , Fasting , Gene Expression Regulation , Insulin/biosynthesis , Insulin/metabolism , Insulin Secretion , Intestinal Mucosa/metabolism , Intestines/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Microtomy , Oxyntomodulin/biosynthesis , Oxyntomodulin/genetics , Oxyntomodulin/metabolism , Period Circadian Proteins/metabolism , Photoperiod , Signal Transduction , Suprachiasmatic Nucleus/drug effects , Suprachiasmatic Nucleus/physiology , Tissue Culture Techniques
17.
J Endocrinol ; 224(2): 139-47, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25385872

ABSTRACT

Clinical cases of glucocorticoid (GC) excess are characterized by increased fat mass and obesity through the accumulation of white adipocytes. The effects of GCs on growth and function of brown adipose tissue are unknown and may contribute to the negative energy balance observed clinically. This study aims to evaluate the effect of GCs on proliferation, differentiation, and metabolic function of brown adipocytes. Human brown adipocytes sourced from supraclavicular fat biopsies were grown in culture and differentiated to mature adipocytes. Human white adipocytes sourced from subcutaneous abdominal fat biopsies were cultured as controls. Effects of dexamethasone on growth, differentiation (UCP1, CIDEA, and PPARGC1A expression), and function (oxygen consumption rate (OCR)) of brown adipocytes were quantified. Dexamethasone (1 µM) significantly stimulated the proliferation of brown preadipocytes and reduced that of white preadipocytes. During differentiation, dexamethasone (at 0.1, 1, and 10 µM) stimulated the expression of UCP1, CIDEA, and PPARGC1A in a concentration-dependent manner and enhanced by fourfold to sixfold the OCR of brown adipocytes. Isoprenaline (100 nM) significantly increased (P<0.05) expression of UCP1 and OCR of brown adipocytes. These effects were significantly reduced (P<0.05) by dexamethasone. Thus, we show that dexamethasone stimulates the proliferation, differentiation, and function of human brown adipocytes but inhibits adrenergic stimulation of the functioning of brown adipocytes. We conclude that GCs exert complex effects on development and function of brown adipocytes. These findings provide strong evidence for an effect of GCs on the biology of human brown adipose tissue (BAT) and for the involvement of the BAT system in the metabolic manifestation of Cushing's syndrome.


Subject(s)
Adipocytes, Brown/drug effects , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Adipocytes, Brown/physiology , Adrenergic Neurons/drug effects , Adrenergic Neurons/metabolism , Adult , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Female , Gene Expression Regulation/drug effects , Humans , Ion Channels/genetics , Ion Channels/metabolism , Male , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Oxygen Consumption/drug effects , Oxygen Consumption/genetics , Uncoupling Protein 1
18.
Physiol Rep ; 2(11)2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25413316

ABSTRACT

PET-CT using (18)F-FDG is employed for detecting brown adipose tissue (BAT) in humans. Alternative methods are needed because of the radiation and cost of PET-CT imaging. The aim was to evaluate the accuracy of infrared thermography (IRT) in detecting human BAT benchmarked to PET-CT imaging. Seventeen individuals underwent a total of 29 PET-CT scans, 12 of whom were studied twice, after 2 h of cold stimulation at 19°C, in parallel with measurement of skin temperatures overlying the supraclavicular (SCV) fossa and the lateral upper chest (control), before and after cold stimulation. Of the 29 scans, 20 were BAT positive after cold stimulation. The mean left SCV temperature tended to be higher in the BAT-positive group before and during cooling. It was significantly higher (P = 0.04) than the temperature of the control area, which fell significantly during cooling in the BAT-positive (-1.2 ± 0.3°C, P = 0.002) but not in the negative (-0.2 ± 0.4°C) group. The temperature difference (Δtemp) between left SCV and chest increased during cooling in the BAT-positive (1.2 ± 0.2 to 2.0 ± 0.3°C, P < 0.002) but not in the negative group (0.6 ± 0.1 to 0.7 ± 0.1°C). A Δtemp of 0.9°C conferred a positive predictive value of 85% for SCV BAT, superior to that of SCV temperature. The findings were similar on the right. In conclusion, the Δtemp is significantly and consistently greater in BAT-positive subjects. The Δtemp quantified by IRT after 2-h cooling shows promise as a noninvasive convenient technique for studying SCV BAT function.

19.
J Mol Endocrinol ; 52(1): R1-16, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23997239

ABSTRACT

In most species, endogenous circadian clocks regulate 24-h rhythms of behavior and physiology. Clock disruption has been associated with decreased cognitive performance and increased propensity to develop obesity, diabetes, and cancer. Many hormonal factors show robust diurnal secretion rhythms, some of which are involved in mediating clock output from the brain to peripheral tissues. In this review, we describe the mechanisms of clock-hormone interaction in mammals, the contribution of different tissue oscillators to hormonal regulation, and how changes in circadian timing impinge on endocrine signalling and downstream processes. We further summarize recent findings suggesting that hormonal signals may feed back on circadian regulation and how this crosstalk interferes with physiological and metabolic homeostasis.


Subject(s)
Circadian Rhythm/physiology , Endocrine System/physiology , Adrenal Glands/physiology , Animals , Circadian Clocks/physiology , Hormones/physiology , Humans , Pineal Gland/physiology , Suprachiasmatic Nucleus/physiology
20.
Am J Physiol Endocrinol Metab ; 304(10): E1053-63, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23531614

ABSTRACT

Perturbation of circadian rhythmicity in mammals, either by environmental influences such as shiftwork or by genetic manipulation, has been associated with metabolic disturbance and the development of obesity and diabetes. Circadian clocks are based on transcriptional/translational feedback loops, comprising positive and negative components. Whereas the metabolic effects of deletion of the positive arm of the clock gene machinery, as in Clock- or Bmal1-deficient mice, have been well characterized, inactivation of Period genes (Per1-3) as components of the negative arm have more complex, sometimes contradictory effects on energy homeostasis. The CRYPTOCHROMEs are critical interaction partners of PERs, and simultaneous deletion of Cry1 and -2 results in behavioral and molecular circadian arrhythmicity. We show that, when challenged with a high-fat diet, Cry1/2(-/-) mice rapidly gain weight and surpass that of wild-type mice, despite displaying hypophagia. Transcript analysis of white adipose tissue reveals upregulated expression of lipogenic genes, many of which are insulin targets. High-fat diet-induced hyperinsulinemia, as a result of potentiated insulin secretion, coupled with selective insulin sensitivity in adipose tissue of Cry1/2(-/-) mice, correlates with increased lipid uptake. Collectively, these data indicate that Cry deficiency results in an increased vulnerability to high-fat diet-induced obesity that might be mediated by increased insulin secretion and lipid storage in adipose tissues.


Subject(s)
Adipose Tissue, White/metabolism , Circadian Rhythm/physiology , Cryptochromes/physiology , Hyperinsulinism/metabolism , Insulin Resistance/physiology , Animals , Blood Glucose/metabolism , Calorimetry, Indirect/methods , Circadian Rhythm/genetics , Cryptochromes/genetics , Diet, High-Fat , Histocytochemistry , Hyperinsulinism/etiology , Hyperinsulinism/genetics , Insulin/blood , Insulin Resistance/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/chemistry , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Up-Regulation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...