Browse

You are looking at 121 - 130 of 198 items for

  • Refine by access: Open Access content only x
Clear All
David M Golding School of Biosciences, Cardiff University, Cardiff, UK

Search for other papers by David M Golding in
Google Scholar
PubMed
Close
,
Daniel J Rees Institute of Life Sciences, College of Medicine, Swansea University, Swansea, UK

Search for other papers by Daniel J Rees in
Google Scholar
PubMed
Close
,
Jennifer R Davies Behavioural Genetics Group, MRC Centre for Neuropsychiatric Genetics and Genomics, Neuroscience and Mental Health Research Institute, Schools of Medicine & Psychology, Cardiff University, Cardiff, UK

Search for other papers by Jennifer R Davies in
Google Scholar
PubMed
Close
,
Dinko Relkovic Behavioural Genetics Group, MRC Centre for Neuropsychiatric Genetics and Genomics, Neuroscience and Mental Health Research Institute, Schools of Medicine & Psychology, Cardiff University, Cardiff, UK

Search for other papers by Dinko Relkovic in
Google Scholar
PubMed
Close
,
Hannah V Furby Behavioural Genetics Group, MRC Centre for Neuropsychiatric Genetics and Genomics, Neuroscience and Mental Health Research Institute, Schools of Medicine & Psychology, Cardiff University, Cardiff, UK

Search for other papers by Hannah V Furby in
Google Scholar
PubMed
Close
,
Irina A Guschina School of Biosciences, Cardiff University, Cardiff, UK

Search for other papers by Irina A Guschina in
Google Scholar
PubMed
Close
,
Anna L Hopkins School of Biosciences, Cardiff University, Cardiff, UK

Search for other papers by Anna L Hopkins in
Google Scholar
PubMed
Close
,
Jeffrey S Davies Institute of Life Sciences, College of Medicine, Swansea University, Swansea, UK

Search for other papers by Jeffrey S Davies in
Google Scholar
PubMed
Close
,
James L Resnick Center for Mammalian Genetics, University of Florida, College of Medicine, Gainesville, Florida, USA

Search for other papers by James L Resnick in
Google Scholar
PubMed
Close
,
Anthony R Isles Behavioural Genetics Group, MRC Centre for Neuropsychiatric Genetics and Genomics, Neuroscience and Mental Health Research Institute, Schools of Medicine & Psychology, Cardiff University, Cardiff, UK

Search for other papers by Anthony R Isles in
Google Scholar
PubMed
Close
, and
Timothy Wells School of Biosciences, Cardiff University, Cardiff, UK

Search for other papers by Timothy Wells in
Google Scholar
PubMed
Close

Prader–Willi syndrome (PWS), a neurodevelopmental disorder caused by loss of paternal gene expression from 15q11–q13, is characterised by growth retardation, hyperphagia and obesity. However, as single gene mutation mouse models for this condition display an incomplete spectrum of the PWS phenotype, we have characterised the metabolic impairment in a mouse model for ‘full’ PWS, in which deletion of the imprinting centre (IC) abolishes paternal gene expression from the entire PWS cluster. We show that PWS-IC del mice displayed postnatal growth retardation, with reduced body weight, hyperghrelinaemia and marked abdominal leanness; proportionate retroperitoneal, epididymal/omental and inguinal white adipose tissue (WAT) weights being reduced by 82%, 84% and 67%, respectively. PWS-IC del mice also displayed a 48% reduction in proportionate interscapular brown adipose tissue (isBAT) weight with significant ‘beiging’ of abdominal WAT, and a 2°C increase in interscapular surface body temperature. Maintenance of PWS-IC del mice under thermoneutral conditions (30°C) suppressed the thermogenic activity in PWS-IC del males, but failed to elevate the abdominal WAT weight, possibly due to a normalisation of caloric intake. Interestingly, PWS-IC del mice also showed exaggerated food hoarding behaviour with standard and high-fat diets, but despite becoming hyperphagic when switched to a high-fat diet, PWS-IC del mice failed to gain weight. This evidence indicates that, unlike humans with PWS, loss of paternal gene expression from the PWS cluster in mice results in abdominal leanness. Although reduced subcutaneous insulation may lead to exaggerated heat loss and thermogenesis, abdominal leanness is likely to arise from a reduced lipid storage capacity rather than increased energy utilisation in BAT.

Open access
Denys deCatanzaro Department of Psychology, Neuroscience & Behaviour, McMaster University, Hamilton, Ontario, Canada

Search for other papers by Denys deCatanzaro in
Google Scholar
PubMed
Close
and
Tyler Pollock Department of Psychology, Neuroscience & Behaviour, McMaster University, Hamilton, Ontario, Canada

Search for other papers by Tyler Pollock in
Google Scholar
PubMed
Close

Estradiol-17β (E2) plays critical roles in female maturation, sexual receptivity, ovulation and fertility. In many mammals, contact with males can similarly affect these female parameters, whereas male excretions contain significant quantities of E2. We administered radiolabeled estradiol ([3H]E2) to male mice in doses representing a small fraction of their endogenous E2. These males were paired with sexually receptive females, and radioactivity was traced into the females’ systems. In Experiment 1, males were given [3H]E2 at 24 and 1 h before mating. Male-to-female [3H]E2 transfer intensified with increasing numbers of intromissions and spiked in the uterus after insemination. In Experiment 2, sexually experienced young males received [3H]E2 at 72 and 24 h before mating, and all mated to ejaculation. The copulatory plug deposited in the female reproductive tract contained substantial levels of radioactivity. The uteri, other tissues and blood serum of females displayed radioactivity indicative of E2 transfer. In Experiment 3, radioactivity was observed 3 and 18 h after insemination in the females’ uteri and other tissues, including parts of the brain. In Experiment 4, we observed substantial levels of radioactivity in semen as well as the copulatory plugs retrieved from the females after mating. Transferred E2 could directly affect abundant estrogen receptors in the female reproductive tract without potential metabolism by the liver. Sexually transferred E2 may facilitate uterine preparation for blastocyst implantation. These data converge with several lines of evidence indicating that male-sourced E2 can transfer to proximate females in bioactive form, contributing to various mammalian pheromonal effects.

Open access
Bernard Freudenthal Molecular Endocrinology Laboratory, Department of Medicine, Imperial College London, London, UK

Search for other papers by Bernard Freudenthal in
Google Scholar
PubMed
Close
,
John Logan Molecular Endocrinology Laboratory, Department of Medicine, Imperial College London, London, UK

Search for other papers by John Logan in
Google Scholar
PubMed
Close
,
Sanger Institute Mouse Pipelines Mouse Pipelines, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK

Search for other papers by Sanger Institute Mouse Pipelines in
Google Scholar
PubMed
Close
,
Peter I Croucher Garvan Institute of Medical Research, Sydney, New South Wales, Australia

Search for other papers by Peter I Croucher in
Google Scholar
PubMed
Close
,
Graham R Williams Molecular Endocrinology Laboratory, Department of Medicine, Imperial College London, London, UK

Search for other papers by Graham R Williams in
Google Scholar
PubMed
Close
, and
J H Duncan Bassett Molecular Endocrinology Laboratory, Department of Medicine, Imperial College London, London, UK

Search for other papers by J H Duncan Bassett in
Google Scholar
PubMed
Close

The genetic determinants of osteoporosis remain poorly understood, and there is a large unmet need for new treatments in our ageing society. Thus, new approaches for gene discovery in skeletal disease are required to complement the current genome-wide association studies in human populations. The International Knockout Mouse Consortium (IKMC) and the International Mouse Phenotyping Consortium (IMPC) provide such an opportunity. The IKMC generates knockout mice representing each of the known protein-coding genes in C57BL/6 mice and, as part of the IMPC initiative, the Origins of Bone and Cartilage Disease project identifies mutants with significant outlier skeletal phenotypes. This initiative will add value to data from large human cohorts and provide a new understanding of bone and cartilage pathophysiology, ultimately leading to the identification of novel drug targets for the treatment of skeletal disease.

Open access
Sushil K Mahata VA San Diego Healthcare System Metabolic Physiology & Ultrastructural Biology Lab., Department of Medicine, University of California at San Diego, La Jolla, CA, USA

Search for other papers by Sushil K Mahata in
Google Scholar
PubMed
Close
,
Hong Zheng Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA

Search for other papers by Hong Zheng in
Google Scholar
PubMed
Close
,
Sumana Mahata Caltech Division of Biology, California Institute of Technology, Pasadena, CA, USA

Search for other papers by Sumana Mahata in
Google Scholar
PubMed
Close
,
Xuefei Liu Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA

Search for other papers by Xuefei Liu in
Google Scholar
PubMed
Close
, and
Kaushik P Patel Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA

Search for other papers by Kaushik P Patel in
Google Scholar
PubMed
Close

One of the key mechanisms involved in sympathoexcitation in chronic heart failure (HF) is the activation of the adrenal glands. Impact of the elevated catecholamines on the hemodynamic parameters has been previously demonstrated. However, studies linking the structural effects of such overactivation with secretory performance and cell metabolism in the adrenomedullary chromaffin cells in vivo have not been previously reported. In this study, HF was induced in male Sprague-Dawley rats by ligation of the left coronary artery. Five weeks after surgery, cardiac function was assessed by ventricular hemodynamics. HF rats showed increased adrenal weight and adrenal catecholamine levels (norepinephrine, epinephrine and dopamine) compared with sham-operated rats. Rats with HF demonstrated increased small synaptic and dense core vesicle in splanchnic–adrenal synapses indicating trans-synaptic activation of catecholamine biosynthetic enzymes, increased endoplasmic reticulum and Golgi lumen width to meet the demand of increased catecholamine synthesis and release, and more mitochondria with dilated cristae and glycogen to accommodate for the increased energy demand for the increased biogenesis and exocytosis of catecholamines from the adrenal medulla. These findings suggest that increased trans-synaptic activation of the chromaffin cells within the adrenal medulla may lead to increased catecholamines in the circulation which in turn contributes to the enhanced neurohumoral drive, providing a unique mechanistic insight for enhanced catecholamine levels in plasma commonly observed in chronic HF condition.

Open access
K S Wilson The University/BHF Centre for Cardiovascular Science, University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK

Search for other papers by K S Wilson in
Google Scholar
PubMed
Close
,
C S Tucker The University/BHF Centre for Cardiovascular Science, University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK

Search for other papers by C S Tucker in
Google Scholar
PubMed
Close
,
E A S Al-Dujaili The University/BHF Centre for Cardiovascular Science, University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK

Search for other papers by E A S Al-Dujaili in
Google Scholar
PubMed
Close
,
M C Holmes The University/BHF Centre for Cardiovascular Science, University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK

Search for other papers by M C Holmes in
Google Scholar
PubMed
Close
,
P W F Hadoke The University/BHF Centre for Cardiovascular Science, University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK

Search for other papers by P W F Hadoke in
Google Scholar
PubMed
Close
,
C J Kenyon The University/BHF Centre for Cardiovascular Science, University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK

Search for other papers by C J Kenyon in
Google Scholar
PubMed
Close
, and
M A Denvir The University/BHF Centre for Cardiovascular Science, University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, UK

Search for other papers by M A Denvir in
Google Scholar
PubMed
Close

Glucocorticoids (GCs) in utero influence embryonic development with consequent programmed effects on adult physiology and pathophysiology and altered susceptibility to cardiovascular disease. However, in viviparous species, studies of these processes are compromised by secondary maternal influences. The zebrafish, being fertilised externally, avoids this problem and has been used here to investigate the effects of transient alterations in GC activity during early development. Embryonic fish were treated either with dexamethasone (a synthetic GC), an antisense GC receptor (GR) morpholino (GR Mo), or hypoxia for the first 120h post fertilisation (hpf); responses were measured during embryonic treatment or later, post treatment, in adults. All treatments reduced cortisol levels in embryonic fish to similar levels. However, morpholino- and hypoxia-treated embryos showed delayed physical development (slower hatching and straightening of head–trunk angle, shorter body length), less locomotor activity, reduced tactile responses and anxiogenic activity. In contrast, dexamethasone-treated embryos showed advanced development and thigmotaxis but no change in locomotor activity or tactile responses. Gene expression changes were consistent with increased (dexamethasone) and decreased (hypoxia, GR Mo) GC activity. In adults, stressed cortisol values were increased with dexamethasone and decreased by GR Mo and hypoxia pre-treatments. Other responses were similarly differentially affected. In three separate tests of behaviour, dexamethasone-programmed fish appeared ‘bolder’ than matched controls, whereas Mo and hypoxia pre-treated fish were unaffected or more reserved. Similarly, the dexamethasone group but not the Mo or hypoxia groups were heavier, longer and had a greater girth than controls. Hyperglycaemia and expression of GC responsive gene (pepck) were also increased in the dexamethasone group. We conclude that GC activity controls many aspects of early-life growth and development in the zebrafish and that, like other species, manipulating GC status pharmacologically, physiologically or genetically in early life leads to programmable metabolic and behavioural traits in adulthood.

Open access
T V Novoselova Centre for Endocrinology, Queen Mary University of London, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Charterhouse Square, London, UK

Search for other papers by T V Novoselova in
Google Scholar
PubMed
Close
,
R Larder University of Cambridge Metabolic Research Laboratories, MRC Metabolic Disease Unit, Wellcome Trust-MRC Institute of Metabolic Science and NIHR Cambridge Biomedical Research Centre, Addenbrooke’s Hospital, Cambridge, UK

Search for other papers by R Larder in
Google Scholar
PubMed
Close
,
D Rimmington University of Cambridge Metabolic Research Laboratories, MRC Metabolic Disease Unit, Wellcome Trust-MRC Institute of Metabolic Science and NIHR Cambridge Biomedical Research Centre, Addenbrooke’s Hospital, Cambridge, UK

Search for other papers by D Rimmington in
Google Scholar
PubMed
Close
,
C Lelliott Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK

Search for other papers by C Lelliott in
Google Scholar
PubMed
Close
,
E H Wynn Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK

Search for other papers by E H Wynn in
Google Scholar
PubMed
Close
,
R J Gorrigan Centre for Endocrinology, Queen Mary University of London, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Charterhouse Square, London, UK

Search for other papers by R J Gorrigan in
Google Scholar
PubMed
Close
,
P H Tate Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK

Search for other papers by P H Tate in
Google Scholar
PubMed
Close
,
L Guasti Centre for Endocrinology, Queen Mary University of London, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Charterhouse Square, London, UK

Search for other papers by L Guasti in
Google Scholar
PubMed
Close
,
The Sanger Mouse Genetics Project Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK

Search for other papers by The Sanger Mouse Genetics Project in
Google Scholar
PubMed
Close
,
S O’Rahilly University of Cambridge Metabolic Research Laboratories, MRC Metabolic Disease Unit, Wellcome Trust-MRC Institute of Metabolic Science and NIHR Cambridge Biomedical Research Centre, Addenbrooke’s Hospital, Cambridge, UK

Search for other papers by S O’Rahilly in
Google Scholar
PubMed
Close
,
A J L Clark Centre for Endocrinology, Queen Mary University of London, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Charterhouse Square, London, UK

Search for other papers by A J L Clark in
Google Scholar
PubMed
Close
,
D W Logan Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK

Search for other papers by D W Logan in
Google Scholar
PubMed
Close
,
A P Coll University of Cambridge Metabolic Research Laboratories, MRC Metabolic Disease Unit, Wellcome Trust-MRC Institute of Metabolic Science and NIHR Cambridge Biomedical Research Centre, Addenbrooke’s Hospital, Cambridge, UK

Search for other papers by A P Coll in
Google Scholar
PubMed
Close
, and
L F Chan Centre for Endocrinology, Queen Mary University of London, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Charterhouse Square, London, UK

Search for other papers by L F Chan in
Google Scholar
PubMed
Close

Melanocortin receptor accessory protein 2 (MRAP2) is a transmembrane accessory protein predominantly expressed in the brain. Both global and brain-specific deletion of Mrap2 in mice results in severe obesity. Loss-of-function MRAP2 mutations have also been associated with obesity in humans. Although MRAP2 has been shown to interact with MC4R, a G protein-coupled receptor with an established role in energy homeostasis, appetite regulation and lipid metabolism, the mechanisms through which loss of MRAP2 causes obesity remains uncertain. In this study, we used two independently derived lines of Mrap2 deficient mice (Mrap2 tm1a/tm1a ) to further study the role of Mrap2 in the regulation of energy balance and peripheral lipid metabolism. Mrap2 tm1a/tm1a mice have a significant increase in body weight, with increased fat and lean mass, but without detectable changes in food intake or energy expenditure. Transcriptomic analysis showed significantly decreased expression of Sim1, Trh, Oxt and Crh within the hypothalamic paraventricular nucleus of Mrap2 tm1a/tm1a mice. Circulating levels of both high-density lipoprotein and low-density lipoprotein were significantly increased in Mrap2 deficient mice. Taken together, these data corroborate the role of MRAP2 in metabolic regulation and indicate that, at least in part, this may be due to defective central melanocortin signalling.

Open access
Geoffrey L Hammond Departments of Cellular & Physiological Sciences and Obstetrics & Gynaecology, University of British Columbia, Vancouver, British Columbia, Canada

Search for other papers by Geoffrey L Hammond in
Google Scholar
PubMed
Close

Biologically active steroids are transported in the blood by albumin, sex hormone-binding globulin (SHBG), and corticosteroid-binding globulin (CBG). These plasma proteins also regulate the non-protein-bound or ‘free’ fractions of circulating steroid hormones that are considered to be biologically active; as such, they can be viewed as the ‘primary gatekeepers of steroid action’. Albumin binds steroids with limited specificity and low affinity, but its high concentration in blood buffers major fluctuations in steroid concentrations and their free fractions. By contrast, SHBG and CBG play much more dynamic roles in controlling steroid access to target tissues and cells. They bind steroids with high (~nM) affinity and specificity, with SHBG binding androgens and estrogens and CBG binding glucocorticoids and progesterone. Both are glycoproteins that are structurally unrelated, and they function in different ways that extend beyond their transportation or buffering functions in the blood. Plasma SHBG and CBG production by the liver varies during development and different physiological or pathophysiological conditions, and abnormalities in the plasma levels of SHBG and CBG or their abilities to bind steroids are associated with a variety of pathologies. Understanding how the unique structures of SHBG and CBG determine their specialized functions, how changes in their plasma levels are controlled, and how they function outside the blood circulation provides insight into how they control the freedom of steroids to act in health and disease.

Open access
Ya-Li Yang Research Laboratory for Reproductive Health, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China

Search for other papers by Ya-Li Yang in
Google Scholar
PubMed
Close
,
Li-Rong Ren Shenzhen Key Laboratory of Birth Defects, Shenzhen Baoan Maternal and Child Health Hospital, Shenzhen, Guangdong, China

Search for other papers by Li-Rong Ren in
Google Scholar
PubMed
Close
,
Li-Feng Sun Research Laboratory for Reproductive Health, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China

Search for other papers by Li-Feng Sun in
Google Scholar
PubMed
Close
,
Chen Huang Research Laboratory for Reproductive Health, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
University of Chinese Academy of Sciences, Shenzhen, China

Search for other papers by Chen Huang in
Google Scholar
PubMed
Close
,
Tian-Xia Xiao Research Laboratory for Reproductive Health, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China

Search for other papers by Tian-Xia Xiao in
Google Scholar
PubMed
Close
,
Bao-Bei Wang Research Laboratory for Reproductive Health, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China

Search for other papers by Bao-Bei Wang in
Google Scholar
PubMed
Close
,
Jie Chen Research Laboratory for Reproductive Health, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China

Search for other papers by Jie Chen in
Google Scholar
PubMed
Close
,
Brian A Zabel Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, California, USA, and Center for Molecular Biology and Medicine, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA

Search for other papers by Brian A Zabel in
Google Scholar
PubMed
Close
,
Peigen Ren Research Laboratory for Reproductive Health, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China

Search for other papers by Peigen Ren in
Google Scholar
PubMed
Close
, and
Jian V Zhang Research Laboratory for Reproductive Health, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China

Search for other papers by Jian V Zhang in
Google Scholar
PubMed
Close

Chemerin, a chemokine, plays important roles in immune responses, inflammation, adipogenesis, and carbohydrate metabolism. Our recent research has shown that chemerin has an inhibitory effect on hormone secretion from the testis and ovary. However, whether G protein-coupled receptor 1 (GPR1), the active receptor for chemerin, regulates steroidogenesis and luteolysis in the corpus luteum is still unknown. In this study, we established a pregnant mare serum gonadotropin-human chorionic gonadotropin (PMSG-hCG) superovulation model, a prostaglandin F2α (PGF2α) luteolysis model, and follicle and corpus luteum culture models to analyze the role of chemerin signaling through GPR1 in the synthesis and secretion of gonadal hormones during follicular/luteal development and luteolysis. Our results, for the first time, show that chemerin and GPR1 are both differentially expressed in the ovary over the course of the estrous cycle, with highest levels in estrus and metestrus. GPR1 has been localized to granulosa cells, cumulus cells, and the corpus luteum by immunohistochemistry (IHC). In vitro, we found that chemerin suppresses hCG-induced progesterone production in cultured follicle and corpus luteum and that this effect is attenuated significantly by anti-GPR1 MAB treatment. Furthermore, when the phosphoinositide 3-kinase (PI3K) pathway was blocked, the attenuating effect of GPR1 MAB was abrogated. Interestingly, PGF2α induces luteolysis through activation of caspase-3, leading to a reduction in progesterone secretion. Treatment with GPR1 MAB blocked the PGF2α effect on caspase-3 expression and progesterone secretion. This study indicates that chemerin/GPR1 signaling directly or indirectly regulates progesterone synthesis and secretion during the processes of follicular development, corpus luteum formation, and PGF2α-induced luteolysis.

Open access
Mark E Cleasby Department of Comparative Biomedical Sciences, Royal Veterinary College, University of London, London, UK

Search for other papers by Mark E Cleasby in
Google Scholar
PubMed
Close
,
Pauline M Jamieson Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK

Search for other papers by Pauline M Jamieson in
Google Scholar
PubMed
Close
, and
Philip J Atherton Division of Medical Sciences and Graduate Entry Medicine, University of Nottingham, Medical School, Royal Derby Hospital, Derby, UK

Search for other papers by Philip J Atherton in
Google Scholar
PubMed
Close

Insulin resistance (IR) in skeletal muscle is a key defect mediating the link between obesity and type 2 diabetes, a disease that typically affects people in later life. Sarcopenia (age-related loss of muscle mass and quality) is a risk factor for a number of frailty-related conditions that occur in the elderly. In addition, a syndrome of ‘sarcopenic obesity’ (SO) is now increasingly recognised, which is common in older people and is applied to individuals that simultaneously show obesity, IR and sarcopenia. Such individuals are at an increased risk of adverse health events compared with those who are obese or sarcopenic alone. However, there are no licenced treatments for sarcopenia or SO, the syndrome is poorly defined clinically and the mechanisms that might explain a common aetiology are not yet well characterised. In this review, we detail the nature and extent of the clinical syndrome, highlight some of the key physiological processes that are dysregulated and discuss some candidate molecular pathways that could be implicated in both metabolic and anabolic defects in skeletal muscle, with an eye towards future therapeutic options. In particular, the potential roles of Akt/mammalian target of rapamycin signalling, AMP-activated protein kinase, myostatin, urocortins and vitamin D are discussed.

Open access
Nele Cielen Laboratory of Respiratory Diseases, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium

Search for other papers by Nele Cielen in
Google Scholar
PubMed
Close
,
Nele Heulens Laboratory of Respiratory Diseases, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium

Search for other papers by Nele Heulens in
Google Scholar
PubMed
Close
,
Karen Maes Laboratory of Respiratory Diseases, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium

Search for other papers by Karen Maes in
Google Scholar
PubMed
Close
,
Geert Carmeliet Laboratory of Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium

Search for other papers by Geert Carmeliet in
Google Scholar
PubMed
Close
,
Chantal Mathieu Laboratory of Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium

Search for other papers by Chantal Mathieu in
Google Scholar
PubMed
Close
,
Wim Janssens Laboratory of Respiratory Diseases, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium

Search for other papers by Wim Janssens in
Google Scholar
PubMed
Close
, and
Ghislaine Gayan-Ramirez Laboratory of Respiratory Diseases, Department of Clinical and Experimental Medicine, KULeuven, Leuven, Belgium

Search for other papers by Ghislaine Gayan-Ramirez in
Google Scholar
PubMed
Close

Chronic obstructive pulmonary disease (COPD) is associated with skeletal muscle dysfunction. Vitamin D plays an important role in muscle strength and performance in healthy individuals. Vitamin D deficiency is highly prevalent in COPD, but its role in skeletal muscle dysfunction remains unclear. We examined the time-course effect of vitamin D deficiency on limb muscle function in mice with normal or deficient vitamin D serum levels exposed to air or cigarette smoke for 6, 12 or 18 weeks. The synergy of smoking and vitamin D deficiency increased lung inflammation and lung compliance from 6 weeks on with highest emphysema scores observed at 18 weeks. Smoking reduced body and muscle mass of the soleus and extensor digitorum longus (EDL), but did not affect contractility, despite type II atrophy. Vitamin D deficiency did not alter muscle mass but reduced muscle force over time, downregulated vitamin D receptor expression, and increased muscle lipid peroxidation but did not alter actin and myosin expression, fiber dimensions or twitch relaxation time. The combined effect of smoking and vitamin D deficiency did not further deteriorate muscle function but worsened soleus mass loss and EDL fiber atrophy at 18 weeks. We conclude that the synergy of smoking and vitamin D deficiency in contrast to its effect on lung disease, had different, independent but important noxious effects on skeletal muscles in a mouse model of mild COPD.

Open access