This site uses cookies, tags and tracking settings to store information that help give you the very best browsing experience. If you don't change your settings, we'll assume you're happy with this. More information Dismiss this warning

Browse

You are looking at 81 - 90 of 14,371 items for

  • Refine by access: All content x
Clear All
Elizabeth M Simpson School of Agricultural, Environmental and Veterinary Sciences, Faculty of Science and Health, Charles Sturt University, Wagga Wagga, NSW, Australia
Gulbali Institute, Charles Sturt University, Wagga Wagga, NSW, Australia

Search for other papers by Elizabeth M Simpson in
Google Scholar
PubMed
Close
,
Iain J Clarke School of Agriculture Food and Ecosystem Sciences, Faculty of Science, The University of Melbourne, Parkville, VIC, Australia

Search for other papers by Iain J Clarke in
Google Scholar
PubMed
Close
,
Christopher J Scott School of Dentistry and Medical Science, Faculty of Science and Health, Charles Sturt University, Wagga Wagga, NSW, Australia

Search for other papers by Christopher J Scott in
Google Scholar
PubMed
Close
,
Cyril P Stephen School of Agricultural, Environmental and Veterinary Sciences, Faculty of Science and Health, Charles Sturt University, Wagga Wagga, NSW, Australia
Gulbali Institute, Charles Sturt University, Wagga Wagga, NSW, Australia

Search for other papers by Cyril P Stephen in
Google Scholar
PubMed
Close
,
Alexandra Rao School of Agriculture Food and Ecosystem Sciences, Faculty of Science, The University of Melbourne, Parkville, VIC, Australia

Search for other papers by Alexandra Rao in
Google Scholar
PubMed
Close
, and
Allan J Gunn School of Agricultural, Environmental and Veterinary Sciences, Faculty of Science and Health, Charles Sturt University, Wagga Wagga, NSW, Australia
Gulbali Institute, Charles Sturt University, Wagga Wagga, NSW, Australia

Search for other papers by Allan J Gunn in
Google Scholar
PubMed
Close

Our previous studies showed that microinjection into the median eminence of the sheep of glucagon-like peptide- 1 (GLP-1) or its receptor agonist exendin-4 stimulates luteinising hormone (LH) secretion, but it is unknown whether the same effect may be obtained by systemic administration of the same. The present study measured the response in terms of plasma LH concentrations to intravenous (iv) infusion of exendin-4. A preliminary study showed that infusion of 2 mg exendin-4 into ewes produced a greater LH response in the follicular phase of the oestrous cycle than the luteal phase. Accordingly, the main study monitored plasma LH levels in response to either 0.5 mg or 2 mg exendin-4 or vehicle (normal saline) delivered by jugular infusion for 1 h in the follicular phase of the oestrous cycle. Blood samples were collected at 10 min intervals before, during and after infusion. Both doses of exendin-4 increased mean plasma LH concentrations and increased LH peripheral pulse amplitude. There was no effect on inter-pulse interval or timing of the preovulatory LH surge. These doses of exendin-4 did not alter plasma insulin or glucose concentrations. Quantitative PCR of the gastrointestinal tract samples from a population of ewes confirmed the expression of the preproglucagon gene (GCG). Expression increased aborally and was greatest in the rectum. It is concluded that endogenous GLP-1, most likely derived from the hindgut, may act systemically to stimulate LH secretion. The present data suggest that this effect may be obtained with levels of agonist that are lower than those functioning as an incretin.

Open access
Aune Koitmäe Institute of Neuroanatomy, University Medical Center Hamburg, Hamburg, Germany

Search for other papers by Aune Koitmäe in
Google Scholar
PubMed
Close
,
Yannik Karsten Institute of Neuroanatomy, University Medical Center Hamburg, Hamburg, Germany
Department of Genetics and Molecular Biology, Institute of Biology, University of Magdeburg, Magdeburg, Germany

Search for other papers by Yannik Karsten in
Google Scholar
PubMed
Close
,
Xiaoyu Li Institute of Neuroanatomy, University Medical Center Hamburg, Hamburg, Germany
Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Jiangsu, China

Search for other papers by Xiaoyu Li in
Google Scholar
PubMed
Close
,
Fabio Morellini Research Group Behavioral Biology, Center for Molecular Neurobiology, Hamburg, Germany

Search for other papers by Fabio Morellini in
Google Scholar
PubMed
Close
,
Gabriele M Rune Institute of Cell Biology and Neurobiology, Universitätsmedizin Charité Berlin, Berlin, Germany

Search for other papers by Gabriele M Rune in
Google Scholar
PubMed
Close
, and
Roland A Bender Institute of Neuroanatomy, University Medical Center Hamburg, Hamburg, Germany

Search for other papers by Roland A Bender in
Google Scholar
PubMed
Close

Estrogens regulate synaptic properties and influence hippocampus-related learning and memory via estrogen receptors, which include the G-protein-coupled estrogen receptor 1 (GPER1). Studying mice, in which the GPER1 gene is dysfunctional (GPER1-KO), we here provide evidence for sex-specific roles of GPER1 in these processes. GPER1-KO males showed reduced anxiety in the elevated plus maze, whereas the fear response ('freezing') was specifically increased in GPER1-KO females in a contextual fear conditioning paradigm. In the Morris water maze, spatial learning and memory consolidation was impaired by GPER1 deficiency in both sexes. Notably, in the females, spatial learning deficits and the fear response were more pronounced if mice were in a stage of the estrous cycle, in which E2 serum levels are high (proestrus) or rising (diestrus). On the physiological level, excitability at Schaffer collateral synapses in CA1 increased in GPER1-deficient males and in proestrus/diestrus ('E2 high') females, concordant with an increased hippocampal expression of the AMPA-receptor subunit GluA1 in GPER1-KO males and females as compared to wildtype males. Further changes included an augmented early long-term potentiation (E-LTP) maintenance specifically in GPER1-KO females and an increased hippocampal expression of spinophilin in metestrus/estrus ('E2 low') GPER1-KO females. Our findings suggest modulatory and sex-specific functions of GPER1 in the hippocampal network, which reduce rather than increase neuronal excitability. Dysregulation of these functions may underlie sex-specific cognitive deficits or mood disorders.

Restricted access
T’ng Choong Kwok University/BHF Centre for Cardiovascular Science, University of Edinburgh, Queen’s Medical Research Institute, Edinburgh, United Kingdom

Search for other papers by T’ng Choong Kwok in
Google Scholar
PubMed
Close
and
Roland H Stimson University/BHF Centre for Cardiovascular Science, University of Edinburgh, Queen’s Medical Research Institute, Edinburgh, United Kingdom

Search for other papers by Roland H Stimson in
Google Scholar
PubMed
Close

The identification of brown adipose tissue (BAT) as a thermogenic organ in human adults approximately 20 years ago raised the exciting possibility of activating this tissue as a new treatment for obesity and cardiometabolic disease. [18F]Fluoro-2-deoxyglucose (18F-FDG) combined positron emission tomography and computed tomography (PET/CT) scanning is the most commonly used imaging modality to detect and quantify human BAT activity in vivo. This technique exploits the substantial glucose uptake by BAT during thermogenesis as a marker for BAT metabolism. 18F-FDG PET has provided substantial insights into human BAT physiology, including its regulatory pathways and the effect of obesity and cardiometabolic disease on BAT function. The use of alternative PET tracers and the development of novel techniques such as magnetic resonance imaging, supraclavicular skin temperature measurements, contrast-enhanced ultrasound, near-infrared spectroscopy and microdialysis have all added complementary information to improve our understanding of human BAT. However, many questions surrounding BAT physiology remain unanswered, highlighting the need for further research and novel approaches to investigate this tissue. This review critically discusses current techniques to assess human BAT function in vivo, the insights gained from these modalities and their limitations. We also discuss other promising techniques in development that will help dissect the pathways regulating human thermogenesis and determine the therapeutic potential of BAT activation.

Open access
Fan Yang College of Bioengineering, Chongqing University, Chongqing, P. R. China

Search for other papers by Fan Yang in
Google Scholar
PubMed
Close
,
Shuang Zhao College of Bioengineering, Chongqing University, Chongqing, P. R. China

Search for other papers by Shuang Zhao in
Google Scholar
PubMed
Close
,
Pingqing Wang College of Bioengineering, Chongqing University, Chongqing, P. R. China

Search for other papers by Pingqing Wang in
Google Scholar
PubMed
Close
, and
Wei Xiang School of Advanced Agriculture and Bioengineering, Yangtze Normal University, Chongqing, P. R. China

Search for other papers by Wei Xiang in
Google Scholar
PubMed
Close

Reproduction in mammals is an extremely energy-intensive process and is therefore tightly controlled by the body's energy status. Changes in the nutritional status of the body cause fluctuations in the levels of peripheral metabolic hormone signals, such as leptin, insulin, and ghrelin, which provide feedback to the hypothalamus and integrate to coordinate metabolism and fertility. Therefore, to link energy and reproduction, energetic information must be centrally transmitted to gonadotropin-releasing hormone (GnRH) neurons that act as reproductive gating. However, GnRH neurons themselves are rarely directly involved in energy information perception. First, as key factors in the control of GnRH neurons, we describe the direct role of Kisspeptin and Arg-Phe amide-related peptide-3 (RFRP-3) neurons in mediating metabolic signaling. Second, we focused on summarizing the roles of metabolic hormone-sensitive neurons in mediating peripheral energy hormone signaling. Some of these hormone-sensitive neurons can directly transmit energy information to GnRH neurons, such as Orexin neurons, while others act indirectly through other neurons such as Kisspeptin, RFRP-3 neuron, and (pituitary adenylate cyclase-activating polypeptide) PACAP neurons. In addition, as another important aspect of the integration of metabolism and reproduction, the impact of reproductive signaling itself on metabolic function was also considered, as exemplified by our examination of the role of Kisspeptin and RFRP-3 in feeding control. This review summarizes the latest research progress in related fields, in order to more fully understand the central neuropeptide network that integrates energy metabolism and reproduction.

Free access
Kirsty G Pringle School of Biomedical Sciences & Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Newcastle, New South Wales, Australia
Mothers and Babies Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia

Search for other papers by Kirsty G Pringle in
Google Scholar
PubMed
Close
and
Lisa K Philp Australian Prostate Cancer Research Centre - Queensland, Centre for Genomics and Personalised Health & School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Princess Alexandra Hospital, Translational Research Institute, Brisbane, Queensland, Australia

Search for other papers by Lisa K Philp in
Google Scholar
PubMed
Close

Angiotensin-converting enzyme 2 (ACE2) is not only the viral receptor for the novel coronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) but is also classically known as a key carboxypeptidase, which through multiple interacting partners plays vital physiological roles in the heart, kidney, lung, and gastrointestinal tract. An accumulating body of evidence has implicated the dysregulation of ACE2 abundance and activity in the pathophysiology of multiple disease states. ACE2 has recently regained attention due to its evolving role in driving the susceptibility and disease severity of coronavirus disease 2019 (COVID-19). This narrative review outlines the current knowledge of the structure and tissue distribution of ACE2, its role in mediating SARS-CoV-2 cellular entry, its interacting partners, and functions. It also highlights how SARS-CoV-2-mediated dysregulation of membrane-bound and circulating soluble ACE2 during infection plays an important role in the pathogenesis of COVID-19. We explore contemporary evidence for the dysregulation of ACE2 in populations that have emerged as most vulnerable to COVID-19 morbidity and mortality, including the elderly, men, and pregnant women, and draw attention to ACE2 dynamics and discrepancies across the mRNA, protein (membrane-bound and circulating), and activity levels. This review highlights the need for improved understanding of the basic biology of ACE2 in populations vulnerable to COVID-19 to best ensure their clinical management and the appropriate prescription of targeted therapeutics.

Free access
J Cantley School of Medicine, University of Dundee, Dundee, United Kingdom of Great Britain and Northern Ireland

Search for other papers by J Cantley in
Google Scholar
PubMed
Close
,
D L Eizirik ULB Center for Diabetes Research, Université Libre de Bruxelles Faculté de Médecine, Bruxelles, Belgium

Search for other papers by D L Eizirik in
Google Scholar
PubMed
Close
,
E Latres JDRF International, New York, NY, USA

Search for other papers by E Latres in
Google Scholar
PubMed
Close
,
C M Dayan Cardiff University School of Medicine, Cardiff, United Kingdom of Great Britain and Northern Ireland

Search for other papers by C M Dayan in
Google Scholar
PubMed
Close
, and
the JDRF-DiabetesUK-INNODIA-nPOD Stockholm Symposium 2022
Search for other papers by the JDRF-DiabetesUK-INNODIA-nPOD Stockholm Symposium 2022 in
Google Scholar
PubMed
Close
the JDRF-DiabetesUK-INNODIA-nPOD Stockholm Symposium 2022

There is a growing understanding that the early phases of type 1 diabetes (T1D) are characterised by a deleterious dialogue between the pancreatic beta cells and the immune system. This, combined with the urgent need to better translate this growing knowledge into novel therapies, provided the background for the JDRF–DiabetesUK–INNODIA–nPOD symposium entitled ‘Islet cells in human T1D: from recent advances to novel therapies’, which took place in Stockholm, Sweden, in September 2022. We provide in this article an overview of the main themes addressed in the symposium, pointing to both promising conclusions and key unmet needs that remain to be addressed in order to achieve better approaches to prevent or reverse T1D.

Open access
Sarah L Armour Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Denmark

Search for other papers by Sarah L Armour in
Google Scholar
PubMed
Close
,
Jade E Stanley Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA

Search for other papers by Jade E Stanley in
Google Scholar
PubMed
Close
,
James Cantley Division of Cellular and Systems Medicine, School of Medicine, University of Dundee, UK

Search for other papers by James Cantley in
Google Scholar
PubMed
Close
,
E Danielle Dean Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
Division of Diabetes, Endocrinology, & Metabolism, Vanderbilt University Medical Center School of Medicine, Nashville, Tennessee, USA

Search for other papers by E Danielle Dean in
Google Scholar
PubMed
Close
, and
Jakob G Knudsen Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Denmark

Search for other papers by Jakob G Knudsen in
Google Scholar
PubMed
Close

Since the discovery of glucagon 100 years ago, the hormone and the pancreatic islet alpha cells that produce it have remained enigmatic relative to insulin-producing beta cells. Canonically, alpha cells have been described in the context of glucagon’s role in glucose metabolism in liver, with glucose as the primary nutrient signal regulating alpha cell function. However, current data reveal a more holistic model of metabolic signalling, involving glucagon-regulated metabolism of multiple nutrients by the liver and other tissues, including amino acids and lipids, providing reciprocal feedback to regulate glucagon secretion and even alpha cell mass. Here we describe how various nutrients are sensed, transported and metabolised in alpha cells, providing an integrative model for the metabolic regulation of glucagon secretion and action. Importantly, we discuss where these nutrient-sensing pathways intersect to regulate alpha cell function and highlight key areas for future research.

Free access
Jun Yang Centre of Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Victoria, Australia
Department of Medicine, Monash University, Clayton, Victoria, Australia

Search for other papers by Jun Yang in
Google Scholar
PubMed
Close
,
Morag J Young Cardiovascular Endocrinology Laboratory, Discovery & Preclinical Domain, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia

Search for other papers by Morag J Young in
Google Scholar
PubMed
Close
,
Timothy J Cole Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia

Search for other papers by Timothy J Cole in
Google Scholar
PubMed
Close
, and
Peter J Fuller Centre of Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Victoria, Australia

Search for other papers by Peter J Fuller in
Google Scholar
PubMed
Close

Primary aldosteronism, or Conn syndrome, is the most common endocrine cause of hypertension. It is associated with a higher risk of cardiovascular, metabolic and renal diseases, as well as a lower quality of life than for hypertension due to other causes. The multi-systemic effects of primary aldosteronism can be attributed to aldosterone-mediated activation of the mineralocorticoid receptor in a range of tissues. In this review, we explore the signalling pathways of the mineralocorticoid receptor, with a shift from the traditional focus on the regulation of renal sodium–potassium exchange to a broader understanding of its role in the modulation of tissue inflammation, fibrosis and remodelling. The appreciation of primary aldosteronism as a multi-system disease with tissue-specific pathophysiology may lead to more vigilant testing and earlier institution of targeted interventions.

Free access
Leonie Cabot Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Gleueler Straße, Cologne, Germany
Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Straße, Cologne, Germany

Search for other papers by Leonie Cabot in
Google Scholar
PubMed
Close
,
Juliet Erlenbeck-Dinkelmann Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Gleueler Straße, Cologne, Germany

Search for other papers by Juliet Erlenbeck-Dinkelmann in
Google Scholar
PubMed
Close
, and
Henning Fenselau Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Gleueler Straße, Cologne, Germany
Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Straße, Cologne, Germany
Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Straße, Cologne, Germany

Search for other papers by Henning Fenselau in
Google Scholar
PubMed
Close

The brain is tuned to integrate food-derived signals from the gut, allowing it to accurately adjust behavioral and physiological responses in accordance with nutrient availability. A key element of gut-to-brain communication is the relay of neural cues via peripheral sensory neurons (PSN) which harbor functionally specialized peripheral endings innervating the muscular and mucosal layers of gastrointestinal (GI) tract organs. In this review, we detail the properties of GI tract innervating PSN and describe their roles in regulating satiation and glucose metabolism in response to food consumption. We discuss the complex anatomical organization of vagal and spinal PSN subtypes, their peripheral and central projection patterns, and describe the limitations of unselective lesion and ablation approaches to investigate them. We then highlight the recent identification of molecular markers that allow selective targeting of PSN subtypes that innervate GI tract organs. This has facilitated accurately determining their projections, monitoring their responses to gut stimuli, and manipulating their activity. We contend that these recent developments have significantly improved our understanding of PSN-mediated gut-to-brain communication, which may open new therapeutic windows for the treatment of metabolic disorders, such as obesity and type 2 diabetes.

Free access
Romy I Kerbus Department of Anatomy and Centre for Neuroendocrinology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand

Search for other papers by Romy I Kerbus in
Google Scholar
PubMed
Close
,
Megan A Inglis Department of Anatomy and Centre for Neuroendocrinology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand

Search for other papers by Megan A Inglis in
Google Scholar
PubMed
Close
, and
Greg M Anderson Department of Anatomy and Centre for Neuroendocrinology, University of Otago School of Biomedical Sciences, Dunedin, New Zealand

Search for other papers by Greg M Anderson in
Google Scholar
PubMed
Close

Polycystic ovary syndrome (PCOS) is one of the most common causes of infertility in women. Approximately half of the diagnosed individuals also experience the metabolic syndrome. Central and peripheral resistance to the hormones insulin and leptin have been reported to contribute to both metabolic and reproductive dysregulation. In PCOS and preclinical PCOS animal models, circulating insulin and leptin levels are often increased in parallel with the development of hormone resistance; however, it remains uncertain whether these changes contribute to the PCOS state. In this study, we tested whether central actions of protein tyrosine phosphatase 1B (PTP1B) and suppressor of cytokine signaling 3 (SOCS3), negative regulators of insulin and leptin signaling pathways, respectively, play a role in the development of PCOS-like phenotype. A peripubertal dihydrotestosterone (DHT) excess PCOS-like mouse model was used, which exhibits both metabolic and reproductive dysfunction. Mice with knockout of the genes encoding PTP1B and SOCS3 from forebrain neurons were generated, and metabolic and reproductive functions were compared between knockout and control groups. DHT treatment induced mild insulin resistance but not leptin resistance, so the role of SOCS3 could not be tested. As expected, DHT excess abolished estrous cycles and corpora lutea presence and caused increased visceral adiposity and fasting glucose levels. Knockout mice did not show any rescue of reproductive dysfunction but did have reduced adiposity compared to the control DHT mice. These data suggest that negative regulation of central insulin signaling by PTP1B is not responsible for peripubertal DHT excess-induced reproductive impairments but may mediate its increased adiposity effects.

Restricted access