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 1 - 10 of 14,370 items for

  • Refine by access: All content x
Clear All
Elisa Villalobos University/British Heart Foundation Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom

Search for other papers by Elisa Villalobos in
Google Scholar
PubMed
Close
,
Allende Miguelez-Crespo University/British Heart Foundation Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom

Search for other papers by Allende Miguelez-Crespo in
Google Scholar
PubMed
Close
,
Ruth A Morgan University/British Heart Foundation Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
Scotland’s Rural College, The Roslin Institute, Easter Bush Campus, United Kingdom

Search for other papers by Ruth A Morgan in
Google Scholar
PubMed
Close
,
Lisa Ivatt University/British Heart Foundation Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom

Search for other papers by Lisa Ivatt in
Google Scholar
PubMed
Close
,
Mhairi Paul University/British Heart Foundation Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom

Search for other papers by Mhairi Paul in
Google Scholar
PubMed
Close
,
Joanna P Simpson University/British Heart Foundation Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom

Search for other papers by Joanna P Simpson in
Google Scholar
PubMed
Close
,
Natalie Z M Homer University/British Heart Foundation Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom

Search for other papers by Natalie Z M Homer in
Google Scholar
PubMed
Close
,
Dominic Kurian The Roslin Institute, Royal (Dick) School of Veterinary Studies, College of Medicine and Veterinary Medicine, University of Edinburgh, Easter Bush Campus, Edinburgh, United Kingdom

Search for other papers by Dominic Kurian in
Google Scholar
PubMed
Close
,
Judit Aguilar The Roslin Institute, Royal (Dick) School of Veterinary Studies, College of Medicine and Veterinary Medicine, University of Edinburgh, Easter Bush Campus, Edinburgh, United Kingdom

Search for other papers by Judit Aguilar in
Google Scholar
PubMed
Close
,
Rachel A Kline The Roslin Institute, Royal (Dick) School of Veterinary Studies, College of Medicine and Veterinary Medicine, University of Edinburgh, Easter Bush Campus, Edinburgh, United Kingdom

Search for other papers by Rachel A Kline in
Google Scholar
PubMed
Close
,
Thomas M Wishart The Roslin Institute, Royal (Dick) School of Veterinary Studies, College of Medicine and Veterinary Medicine, University of Edinburgh, Easter Bush Campus, Edinburgh, United Kingdom

Search for other papers by Thomas M Wishart in
Google Scholar
PubMed
Close
,
Nicholas M Morton University/British Heart Foundation Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
Centre for Systems Health and Integrated Metabolic Research, Nottingham Trent University, Nottingham, United Kingdom

Search for other papers by Nicholas M Morton in
Google Scholar
PubMed
Close
,
Roland H Stimson University/British Heart Foundation Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom

Search for other papers by Roland H Stimson in
Google Scholar
PubMed
Close
,
Ruth Andrew University/British Heart Foundation Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom

Search for other papers by Ruth Andrew in
Google Scholar
PubMed
Close
,
Brian R Walker University/British Heart Foundation Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom

Search for other papers by Brian R Walker in
Google Scholar
PubMed
Close
, and
Mark Nixon University/British Heart Foundation Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom

Search for other papers by Mark Nixon in
Google Scholar
PubMed
Close

Glucocorticoids modulate glucose homeostasis, acting on metabolically active tissues such as liver, skeletal muscle, and adipose tissue. Intracellular regulation of glucocorticoid action in adipose tissue impacts metabolic responses to obesity. ATP-binding cassette family C member 1 (ABCC1) is a transmembrane glucocorticoid transporter known to limit the accumulation of exogenously administered corticosterone in adipose tissue. However, the role of ABCC1 in the regulation of endogenous glucocorticoid action and its impact on fuel metabolism has not been studied. Here, we investigate the impact of Abcc1 deficiency on glucocorticoid action and high-fat-diet (HFD)-induced obesity. In lean male mice, deficiency of Abcc1 increased endogenous corticosterone levels in skeletal muscle and adipose tissue but did not impact insulin sensitivity. In contrast, Abcc1-deficient male mice on HFD displayed impaired glucose and insulin tolerance, and fasting hyperinsulinaemia, without alterations in tissue corticosterone levels. Proteomics and bulk RNA sequencing revealed that Abcc1 deficiency amplified the transcriptional response to an obesogenic diet in adipose tissue but not in skeletal muscle. Moreover, Abcc1 deficiency impairs key signalling pathways related to glucose metabolism in both skeletal muscle and adipose tissue, in particular those related to OXPHOS machinery and Glut4. Together, our results highlight a role for ABCC1 in regulating glucose homeostasis, demonstrating diet-dependent effects that are not associated with altered tissue glucocorticoid concentrations.

Open access
Andrea Lovdel University/BHF Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh BioQuarter, Edinburgh, UK

Search for other papers by Andrea Lovdel in
Google Scholar
PubMed
Close
,
Karla J Suchacki University/BHF Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh BioQuarter, Edinburgh, UK

Search for other papers by Karla J Suchacki in
Google Scholar
PubMed
Close
,
Fiona Roberts University/BHF Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh BioQuarter, Edinburgh, UK

Search for other papers by Fiona Roberts in
Google Scholar
PubMed
Close
,
Richard J Sulston University/BHF Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh BioQuarter, Edinburgh, UK

Search for other papers by Richard J Sulston in
Google Scholar
PubMed
Close
,
Robert J Wallace Department of Orthopaedics, The University of Edinburgh, Edinburgh, UK

Search for other papers by Robert J Wallace in
Google Scholar
PubMed
Close
,
Benjamin J Thomas University/BHF Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh BioQuarter, Edinburgh, UK

Search for other papers by Benjamin J Thomas in
Google Scholar
PubMed
Close
,
Rachel M B Bell University/BHF Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh BioQuarter, Edinburgh, UK

Search for other papers by Rachel M B Bell in
Google Scholar
PubMed
Close
,
Iris Pruñonosa Cervera University/BHF Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh BioQuarter, Edinburgh, UK

Search for other papers by Iris Pruñonosa Cervera in
Google Scholar
PubMed
Close
,
Gavin J Macpherson Department of Orthopaedic Surgery, Royal Infirmary of Edinburgh, Edinburgh, UK

Search for other papers by Gavin J Macpherson in
Google Scholar
PubMed
Close
,
Nicholas M Morton University/BHF Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh BioQuarter, Edinburgh, UK
Centre for Systems Health and Integrated Metabolic Research, Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, UK

Search for other papers by Nicholas M Morton in
Google Scholar
PubMed
Close
,
Natalie Z M Homer University/BHF Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh BioQuarter, Edinburgh, UK

Search for other papers by Natalie Z M Homer in
Google Scholar
PubMed
Close
,
Karen E Chapman University/BHF Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh BioQuarter, Edinburgh, UK

Search for other papers by Karen E Chapman in
Google Scholar
PubMed
Close
, and
William P Cawthorn University/BHF Centre for Cardiovascular Science, The University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh BioQuarter, Edinburgh, UK

Search for other papers by William P Cawthorn in
Google Scholar
PubMed
Close

Bone marrow adipose tissue (BMAT) comprises >10% of total adipose mass in healthy humans. It increases in diverse conditions, including ageing, obesity, osteoporosis, glucocorticoid therapy, and notably, during caloric restriction (CR). BMAT potentially influences skeletal, metabolic, and immune functions, but the mechanisms of BMAT expansion remain poorly understood. Our hypothesis is that, during CR, excessive glucocorticoid activity drives BMAT expansion. The enzyme 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) amplifies glucocorticoid activity by catalysing intracellular regeneration of active glucocorticoids from inert 11-keto forms. Mice lacking 11β-HSD1 resist metabolic dysregulation and bone loss during exogenous glucocorticoid excess; thus, we hypothesised that 11β-HSD1 knockout mice would also resist excessive glucocorticoid action during CR, thereby restrining BMAT expansion and bone loss. To test this, we first confirmed that 11β-HSD1 is expressed in mouse and human bone marrow. We then investigated the effects of CR in male and female control and 11β-HSD1 knockout mice from 9 to 15 weeks of age. CR increased Hsd11b1 mRNA in adipose tissue and bone marrow. Deletion of Hsd11b1 did not alter bone or BMAT characteristics in mice fed a control diet and had little effect on tibial bone microarchitecture during CR. Notably, Hsd11b1 deletion attenuated the CR-induced increases in BMAT and prevented increases in bone marrow corticosterone in males but not females. This was not associated with suppression of glucocorticoid target genes in bone marrow. Instead, knockout males had increased progesterone in plasma and bone marrow. Together, our findings show that knockout of 11β-HSD1 prevents CR-induced BMAT expansion in a sex-specific manner and highlights progesterone as a potential new regulator of bone marrow adiposity.

Open access
Nawal A Yahya Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, USA

Search for other papers by Nawal A Yahya in
Google Scholar
PubMed
Close
,
Steven R King Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, USA
Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, Texas, USA

Search for other papers by Steven R King in
Google Scholar
PubMed
Close
,
Bo Shi Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, USA

Search for other papers by Bo Shi in
Google Scholar
PubMed
Close
,
Aisha Shaaban Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, USA

Search for other papers by Aisha Shaaban in
Google Scholar
PubMed
Close
,
Nicole E Whitfield Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, USA

Search for other papers by Nicole E Whitfield in
Google Scholar
PubMed
Close
,
Chunmei Yan Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, USA
Department of Obstetrics, The Second Hospital of Shandong University, Jinan, Shandong, People’s Republic of China

Search for other papers by Chunmei Yan in
Google Scholar
PubMed
Close
,
Richard J Kordus Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, USA

Search for other papers by Richard J Kordus in
Google Scholar
PubMed
Close
,
Gail F Whitman-Elia Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, USA

Search for other papers by Gail F Whitman-Elia in
Google Scholar
PubMed
Close
, and
Holly A LaVoie Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, USA

Search for other papers by Holly A LaVoie in
Google Scholar
PubMed
Close

Cells actively engaged in de novo steroidogenesis rely on an expansive intracellular network to efficiently transport cholesterol. The final link in the transport chain is STARD1, which transfers cholesterol to the enzyme complex that initiates steroidogenesis. However, the regulation of ovarian STARD1 is not fully characterized, and even less is known about the upstream cytosolic cholesterol transporters STARD4 and STARD6. Here, we identified both STARD4 and STARD6 mRNAs in the human ovary but only detected STARD4 protein since the primary STARD6 transcript turned out to be a splice variant. Corpora lutea contained the highest levels of STARD4 and STARD1 mRNA and STARD1 protein, while STARD4 protein was uniformly distributed across ovarian tissues. Cyclic AMP analog (8Br-cAMP) and phorbol ester (PMA) individually increased STARD1 and STARD4 mRNA along with STARD1 protein and its phosphoform in cultured primary human luteinized granulosa cells (hGCs). STARD6 transcripts and STARD4 protein were unresponsive to these stimuli. Combining lower doses of PMA and 8Br-cAMP blunted the 8Br-cAMP stimulation of STARD1 protein. Increasing cholesterol levels by blocking its conversion to steroid with aminoglutethimide or by adding LDL reduced the STARD4 mRNA response to stimuli. Sterol depletion reduced the STARD1 mRNA and protein response to PMA. These data support a possible role for STARD4, but not STARD6, in supplying cholesterol for steroidogenesis in the ovary. We demonstrate for the first time how cAMP, PMA and sterol pathways separately and in combination differentially regulate STARD4, STARD6 and STARD1 mRNA levels, as well as STARD1 and STARD4 protein in human primary ovarian cells.

Restricted access
Lorena González Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Instituto de Química y Fisicoquímica Biológicas (UBA-CONICET), Universidad de Buenos Aires, Buenos Aires, Argentina

Search for other papers by Lorena González in
Google Scholar
PubMed
Close
,
Ma Eugenia Díaz Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Instituto de Química y Fisicoquímica Biológicas (UBA-CONICET), Universidad de Buenos Aires, Buenos Aires, Argentina

Search for other papers by Ma Eugenia Díaz in
Google Scholar
PubMed
Close
,
Johanna G Miquet Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Instituto de Química y Fisicoquímica Biológicas (UBA-CONICET), Universidad de Buenos Aires, Buenos Aires, Argentina

Search for other papers by Johanna G Miquet in
Google Scholar
PubMed
Close
,
Ana I Sotelo Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Instituto de Química y Fisicoquímica Biológicas (UBA-CONICET), Universidad de Buenos Aires, Buenos Aires, Argentina

Search for other papers by Ana I Sotelo in
Google Scholar
PubMed
Close
,
Diego Fernández Cátedra de Bioquímica Humana, Facultad de Medicina (UBA), Buenos Aires, Argentina

Search for other papers by Diego Fernández in
Google Scholar
PubMed
Close
,
Fernando P Dominici Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Instituto de Química y Fisicoquímica Biológicas (UBA-CONICET), Universidad de Buenos Aires, Buenos Aires, Argentina

Search for other papers by Fernando P Dominici in
Google Scholar
PubMed
Close
,
Andrzej Bartke Geriatrics Research, Departments of Internal Medicine and Physiology, School of Medicine, Southern Illinois University, Springfield, Illinois, USA

Search for other papers by Andrzej Bartke in
Google Scholar
PubMed
Close
, and
Daniel Turyn Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Instituto de Química y Fisicoquímica Biológicas (UBA-CONICET), Universidad de Buenos Aires, Buenos Aires, Argentina

Search for other papers by Daniel Turyn in
Google Scholar
PubMed
Close
Free access
Michael Merkhassine Loftus Laboratory, Department of Clinical Sciences, Cornell University, College of Veterinary Medicine, Ithaca, New York, USA
VCA Colonial Animal Hospital, Ithaca, New York, USA

Search for other papers by Michael Merkhassine in
Google Scholar
PubMed
Close
,
Reilly W Coch Loftus Laboratory, Department of Clinical Sciences, Cornell University, College of Veterinary Medicine, Ithaca, New York, USA
Weill Cornell College of Medicine, New York, New York, USA

Search for other papers by Reilly W Coch in
Google Scholar
PubMed
Close
,
Carol E Frederick Loftus Laboratory, Department of Clinical Sciences, Cornell University, College of Veterinary Medicine, Ithaca, New York, USA

Search for other papers by Carol E Frederick in
Google Scholar
PubMed
Close
,
Lucinda L Bennett Loftus Laboratory, Department of Clinical Sciences, Cornell University, College of Veterinary Medicine, Ithaca, New York, USA

Search for other papers by Lucinda L Bennett in
Google Scholar
PubMed
Close
,
Seth A Peng Loftus Laboratory, Department of Clinical Sciences, Cornell University, College of Veterinary Medicine, Ithaca, New York, USA
Fate Therapeutics, San Diego, California, USA

Search for other papers by Seth A Peng in
Google Scholar
PubMed
Close
,
Benjamin Morse Loftus Laboratory, Department of Clinical Sciences, Cornell University, College of Veterinary Medicine, Ithaca, New York, USA

Search for other papers by Benjamin Morse in
Google Scholar
PubMed
Close
,
Bethany P Cummings Center for Alimentary and Metabolic Science, Department of Surgery, School of Medicine, University of California, Davis, Sacramento, California, USA
Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA

Search for other papers by Bethany P Cummings in
Google Scholar
PubMed
Close
, and
John P Loftus Loftus Laboratory, Department of Clinical Sciences, Cornell University, College of Veterinary Medicine, Ithaca, New York, USA

Search for other papers by John P Loftus in
Google Scholar
PubMed
Close

Glucagon plays a central role in amino acid (AA) homeostasis. The dog is an established model of glucagon biology, and recently, metabolomic changes in people associated with glucagon infusions have been reported. Glucagon also has effects on the kidney; however, changes in urinary AA concentrations associated with glucagon remain under investigation. Therefore, we aimed to fill these gaps in the canine model by determining the effects of glucagon on the canine plasma metabolome and measuring urine AA concentrations. Employing two constant rate glucagon infusions (CRI) – low-dose (CRI-LO: 3 ng/kg/min) and high-dose (CRI-HI: 50 ng/kg/min) on five research beagles, we monitored interstitial glucose and conducted untargeted liquid chromatography–tandem mass spectrometry (LC-MS/MS) on plasma samples and urine AA concentrations collected pre- and post-infusion. The CRI-HI induced a transient glucose peak (90–120 min), returning near baseline by infusion end, while only the CRI-LO resulted in 372 significantly altered plasma metabolites, primarily reductions (333). Similarly, CRI-HI affected 414 metabolites, with 369 reductions, evidenced by distinct clustering post-infusion via data reduction (PCA and sPLS-DA). CRI-HI notably decreased circulating AA levels, impacting various AA-related and energy-generating metabolic pathways. Urine analysis revealed increased 3-methyl-l-histidine and glutamine, and decreased alanine concentrations post-infusion. These findings demonstrate glucagon’s glucose-independent modulation of the canine plasma metabolome and highlight the dog’s relevance as a translational model for glucagon biology. Understanding these effects contributes to managing dysregulated glucagon conditions and informs treatments impacting glucagon homeostasis.

Open access
Yu Zhou Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
Department of Obstetrics and Gynecology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China

Search for other papers by Yu Zhou in
Google Scholar
PubMed
Close
,
Chao Lian Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China

Search for other papers by Chao Lian in
Google Scholar
PubMed
Close
,
Yingfei Lu Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China

Search for other papers by Yingfei Lu in
Google Scholar
PubMed
Close
,
Tianming Wang Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China

Search for other papers by Tianming Wang in
Google Scholar
PubMed
Close
,
Chengcheng Zhao Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China

Search for other papers by Chengcheng Zhao in
Google Scholar
PubMed
Close
,
Cuilan Zhang Department of Obstetrics and Gynecology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China

Search for other papers by Cuilan Zhang in
Google Scholar
PubMed
Close
,
Min Gong Department of Obstetrics and Gynecology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China

Search for other papers by Min Gong in
Google Scholar
PubMed
Close
,
Jianquan Chen Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
Department of Obstetrics and Gynecology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China

Search for other papers by Jianquan Chen in
Google Scholar
PubMed
Close
, and
Rong Ju Department of Obstetrics and Gynecology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China

Search for other papers by Rong Ju in
Google Scholar
PubMed
Close

Polycystic ovary syndrome (PCOS) is a condition resulting from the interaction between environmental factors and hereditary components, profoundly affecting offspring development. Although the etiology of this disease remains unclear, aberrant in utero androgen exposure is considered one of the pivotal pathogenic factors. Herein, we demonstrate the intergenerational inheritance of PCOS-like phenotypes in F2 female offspring through F1 males caused by maternal testosterone exposure in F0 mice. We found impaired serum hormone expression and reproductive system development in prenatal testosterone-treated F1 male and F2 female mice (PTF1 and PTF2). In addition, downregulated N6-methyladenosine (m6A) methyltransferase and binding proteins induced mRNA hypomethylation in the PTF1 testis, including frizzled-6 (Fzd6). In the PTF2 ovary, decreased FZD6 protein expression inhibited the mammalian target of rapamycin (mTOR) signaling pathway and activated Forkhead box O3 (FoxO3) phosphorylation, which led to impaired follicular development. These data indicate that epigenetic modification of the mTOR signaling pathway could be involved in the intergenerational inheritance of maternal testosterone exposure-induced impairments in the PTF2 ovary through male PTF1 mice.

Restricted access
Xuan Zhou Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou, China

Search for other papers by Xuan Zhou in
Google Scholar
PubMed
Close
,
Yanan Zhang Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou, China

Search for other papers by Yanan Zhang in
Google Scholar
PubMed
Close
,
Youwen Yuan Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou, China

Search for other papers by Youwen Yuan in
Google Scholar
PubMed
Close
,
Fei Teng Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou, China

Search for other papers by Fei Teng in
Google Scholar
PubMed
Close
,
Jiayang Lin Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou, China

Search for other papers by Jiayang Lin in
Google Scholar
PubMed
Close
,
Xueru Ye Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou, China

Search for other papers by Xueru Ye in
Google Scholar
PubMed
Close
,
Yaojin Pan Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou, China

Search for other papers by Yaojin Pan in
Google Scholar
PubMed
Close
, and
Huijie Zhang Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou, China

Search for other papers by Huijie Zhang in
Google Scholar
PubMed
Close

Characteristic symptoms of hyperthyroidism include weight loss, heart palpitation, and sweating. Thyroid hormones (TH) can stimulate thermogenesis through central and peripheral mechanisms. Previous studies have shown an association between dysfunction of cardiotrophin-like cytokine factor 1 (CLCF1) and cold-induced sweating syndrome, with recent research also indicating a link between CLCF1 and brown adipose tissue thermogenesis. However, it remains unclear whether CLCF1 and TH have synergistic or antagonistic effects on thermogenesis. This study aims to investigate the influence of thyroid hormone on circulating CLCF1 levels in humans and explore the potential possibilities of thyroid hormone in regulating energy metabolism by modulating Clcf1 in mice. By recruiting hyperthyroid patients and healthy subjects, we observed significantly lower serum CLCF1 levels in hyperthyroid patients compared to healthy subjects, with serum CLCF1 levels independently associated with hyperthyroidism after adjusting for potential confounders. Tissue analysis from mice treated with T3 revealed a decrease in CLCF1 expression in BAT and iWAT of C57BL/6 mice. These findings suggest that TH may play a role in regulating CLCF1 expression in adipose tissue.

Restricted access
Kaitlyn A Colglazier Lilly Diabetes Center of Excellence, Indiana Biosciences Research Institute, Indianapolis, Indiana, USA

Search for other papers by Kaitlyn A Colglazier in
Google Scholar
PubMed
Close
,
Noyonika Mukherjee Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA

Search for other papers by Noyonika Mukherjee in
Google Scholar
PubMed
Close
,
Christopher J Contreras Division of Endocrinology, Department of Medicine, Roudebush VA Medical Center and Indiana University School of Medicine, Indianapolis, Indiana, USA

Search for other papers by Christopher J Contreras in
Google Scholar
PubMed
Close
, and
Andrew T Templin Lilly Diabetes Center of Excellence, Indiana Biosciences Research Institute, Indianapolis, Indiana, USA
Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
Division of Endocrinology, Department of Medicine, Roudebush VA Medical Center and Indiana University School of Medicine, Indianapolis, Indiana, USA
Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, Indiana, USA

Search for other papers by Andrew T Templin in
Google Scholar
PubMed
Close

β-Cell death contributes to β-cell loss and insulin insufficiency in type 1 diabetes (T1D), and this β-cell demise has been attributed to apoptosis and necrosis. Apoptosis has been viewed as the lone form of programmed β-cell death, and evidence indicates that β-cells also undergo necrosis, regarded as an unregulated or accidental form of cell demise. More recently, studies in non-islet cell types have identified and characterized novel forms of cell death that are biochemically and morphologically distinct from apoptosis and necrosis. Several of these mechanisms of cell death have been categorized as forms of regulated necrosis and linked to inflammation and disease pathogenesis. In this review, we revisit discoveries of β-cell death in humans with diabetes and describe studies characterizing β-cell apoptosis and necrosis. We explore literature on mechanisms of regulated necrosis including necroptosis, ferroptosis and pyroptosis, review emerging literature on the significance of these mechanisms in β-cells, and discuss experimental approaches to differentiate between various mechanisms of β-cell death. Our review of the literature leads us to conclude that more detailed experimental characterization of the mechanisms of β-cell death is warranted, along with studies to better understand the impact of various forms of β-cell demise on islet inflammation and β-cell autoimmunity in pathophysiologically relevant models. Such studies will provide insight into the mechanisms of β-cell loss in T1D and may shed light on new therapeutic approaches to protect β-cells in this disease.

Open access
Renata Risi Department of Experimental Medicine, Sapienza University of Rome, Sapienza University of Rome, Rome, Italy
University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK

Search for other papers by Renata Risi in
Google Scholar
PubMed
Close
,
Antonio Vidal-Puig University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK
Cambridge University Nanjing Centre of Technology and Innovation, Nanjing, P. R. China
Centro de Investigacion Principe Felipe, Valencia, Spain

Search for other papers by Antonio Vidal-Puig in
Google Scholar
PubMed
Close
, and
Guillaume Bidault University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK

Search for other papers by Guillaume Bidault in
Google Scholar
PubMed
Close

Obesity and diabetes represent two increasing and invalidating public health issues that often coexist. It is acknowledged that fat mass excess predisposes to insulin resistance and type 2 diabetes mellitus (T2D), with the increasing incidence of the two diseases significantly associated. Moreover, emerging evidence suggests that obesity might also accelerate the appearance of type 1 diabetes (T1D), which is now a relatively frequent comorbidity in patients with obesity. It is a common clinical finding that not all patients with obesity will develop diabetes at the same level of adiposity, with gender, genetic, and ethnic factors playing an important role in defining the timing of diabetes appearance. The adipose tissue (AT) expandability hypothesis explains this paradigm, indicating that the individual capacity to appropriately store energy surplus in the form of fat within the AT determines and prevents the toxic deposition of lipids in other organs, such as the pancreas. Thus, we posit that when the maximal storing capacity of AT is exceeded, individuals will develop T2D. In this review, we provide insight into mechanisms by which the AT controls pancreas lipid content and homeostasis in case of obesity to offer an adipocentric perspective of pancreatic lipotoxicity in the pathogenesis of diabetes. Moreover, we suggest that improving AT function is a valid therapeutic approach to fighting obesity-associated complications including diabetes.

Open access
Xiong Weng Division of Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee, Scotland, UK

Search for other papers by Xiong Weng in
Google Scholar
PubMed
Close
,
Hao Jiang Gene Expression and Regulation, School of Life Sciences, University of Dundee, Dundee, Scotland, UK

Search for other papers by Hao Jiang in
Google Scholar
PubMed
Close
,
David J Walker Division of Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee, Scotland, UK

Search for other papers by David J Walker in
Google Scholar
PubMed
Close
,
Houjiang Zhou MRC Protein Phosphorylation Unit, School of Life Sciences, Dundee, Scotland, UK

Search for other papers by Houjiang Zhou in
Google Scholar
PubMed
Close
,
De Lin Drug Discovery Unit, School of Life Sciences, University of Dundee, Dundee, Scotland, UK

Search for other papers by De Lin in
Google Scholar
PubMed
Close
,
Jing Wang Science for Life Laboratory, Department of Biomedical and Clinical Sciences (BKV), Linköping University, Linköping, Sweden

Search for other papers by Jing Wang in
Google Scholar
PubMed
Close
, and
Li Kang Division of Cellular and Systems Medicine, School of Medicine, University of Dundee, Dundee, Scotland, UK

Search for other papers by Li Kang in
Google Scholar
PubMed
Close

CD44, a cell surface adhesion receptor and stem cell biomarker, is recently implicated in chronic metabolic diseases. Ablation of CD44 ameliorates adipose tissue inflammation and insulin resistance in obesity. Here, we investigated cell type-specific CD44 expression in human and mouse adipose tissue and further studied how CD44 in preadipocytes regulates adipocyte function. Using Crispr Cas9-mdediated gene deletion and lentivirus-mediated gene re-expression, we discovered that deletion of CD44 promotes adipocyte differentiation and adipogenesis, whereas re-expression of CD44 abolishes this effect and decreases insulin responsiveness and adiponectin secretion in 3T3-L1 cells. Mechanistically, CD44 does so via suppressing Pparg expression. Using quantitative proteomics analysis, we further discovered that cell cycle-regulated pathways were mostly decreased by deletion of CD44. Indeed, re-expression of CD44 moderately restored expression of proteins involved in all phases of the cell cycle. These data were further supported by increased preadipocyte proliferation rates in CD44-deficient cells and re-expression of CD44 diminished this effect. Our data suggest that CD44 plays a crucial role in regulating adipogenesis and adipocyte function possibly through regulating PPARγ and cell cycle-related pathways. This study provides evidence for the first time that CD44 expressed in preadipocytes plays key roles in regulating adipocyte function outside immune cells where CD44 is primarily expressed. Therefore, targeting CD44 in (pre)adipocytes may provide therapeutic potential to treat obesity-associated metabolic complications.

Open access