Browse

You are looking at 11 - 20 of 2,652 items for

  • Refine by access: All content x
Clear All
Junling Wang Gynecologic Department, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China

Search for other papers by Junling Wang in
Google Scholar
PubMed
Close
,
Hongyan Zhang Gynecologic Department, Hainan Traditional Chinese Medicine Hospital, Haikou, China

Search for other papers by Hongyan Zhang in
Google Scholar
PubMed
Close
,
Yue Cao Science and technology department, Guangzhou University of Chinese Medicine, Guangzhou, China

Search for other papers by Yue Cao in
Google Scholar
PubMed
Close
,
Irene Ma Gynecologic Department, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China

Search for other papers by Irene Ma in
Google Scholar
PubMed
Close
,
Xuefang Liang Gynecologic Department, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China

Search for other papers by Xuefang Liang in
Google Scholar
PubMed
Close
, and
Dongfang Xiang Gynecologic Department, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China

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

Postmenopausal osteoporosis (OP) is a prevalent skeletal disease with not fully understood molecular mechanisms. This study aims to investigate the role of circular RNA (circRNA) in postmenopausal OP and to elucidate the potential mechanisms of the circRNA–miRNA–mRNA regulatory network. We obtained circRNA and miRNA expression profiles from postmenopausal OP patients from the Gene Expression Omnibus database. By identifying differentially expressed circRNAs and miRNAs, we constructed a circRNA–miRNA–mRNA network and identified key genes associated with OP. Further, through a range of experimental approaches, including dual-luciferase reporter assays, RNA pull-down experiments, and qRT-PCR, we examined the roles of circ_0134120, miR-590-5p, and STAT3 in the progression of OP. Our findings reveal that the interaction between circ_0134120 and miR-590-5p in regulating STAT3 gene expression is a key mechanism in OP, suggesting the circRNA–miRNA–mRNA network is a potential therapeutic target for this condition.

Restricted access
Abigail R Walker Institute of Reproductive and Developmental Biology, Department Metabolism, Digestion and Reproduction, Imperial College London, London, UK

Search for other papers by Abigail R Walker in
Google Scholar
PubMed
Close
,
Holly A Parkin Institute of Reproductive and Developmental Biology, Department Metabolism, Digestion and Reproduction, Imperial College London, London, UK

Search for other papers by Holly A Parkin in
Google Scholar
PubMed
Close
,
Sung Hye Kim Institute of Reproductive and Developmental Biology, Department Metabolism, Digestion and Reproduction, Imperial College London, London, UK

Search for other papers by Sung Hye Kim in
Google Scholar
PubMed
Close
,
Vasso Terzidou Institute of Reproductive and Developmental Biology, Department Metabolism, Digestion and Reproduction, Imperial College London, London, UK

Search for other papers by Vasso Terzidou in
Google Scholar
PubMed
Close
,
David F Woodward Department of Bioengineering, Imperial College London, London, UK

Search for other papers by David F Woodward in
Google Scholar
PubMed
Close
,
Phillip R Bennett Institute of Reproductive and Developmental Biology, Department Metabolism, Digestion and Reproduction, Imperial College London, London, UK

Search for other papers by Phillip R Bennett in
Google Scholar
PubMed
Close
, and
Aylin C Hanyaloglu Institute of Reproductive and Developmental Biology, Department Metabolism, Digestion and Reproduction, Imperial College London, London, UK

Search for other papers by Aylin C Hanyaloglu in
Google Scholar
PubMed
Close

The prostanoid G protein-coupled receptor (GPCR) EP2 is widely expressed and implicated in endometriosis, osteoporosis, obesity, pre-term labour and cancer. Internalisation and intracellular trafficking are critical for shaping GPCR activity, yet little is known regarding the spatial programming of EP2 signalling and whether this can be exploited pharmacologically. Using three EP2-selective ligands that favour activation of different EP2 pathways, we show that EP2 undergoes limited agonist-driven internalisation but is constitutively internalised via dynamin-dependent, β-arrestin-independent pathways. EP2 was constitutively trafficked to early and very early endosomes (VEE), which was not altered by ligand activation. APPL1, a key adaptor and regulatory protein of the VEE, did not impact EP2 agonist-mediated cAMP. Internalisation was required for ~70% of the acute butaprost- and AH13205-mediated cAMP signalling, yet PGN9856i, a Gαs-biased agonist, was less dependent on receptor internalisation for its cAMP signalling, particularly in human term pregnant myometrial cells that endogenously express EP2. Inhibition of EP2 internalisation partially reduced calcium signalling activated by butaprost or AH13205 and had no effect on PGE2 secretion. This indicates an agonist-dependent differential spatial requirement for Gαs and Gαq/11 signalling and a role for plasma membrane-initiated Gαq/11-Ca2+-mediated PGE2 secretion. These findings reveal a key role for EP2 constitutive internalisation in its signalling and potential spatial bias in mediating its downstream functions. This, in turn, could highlight important considerations for future selective targeting of EP2 signalling pathways.

Open access
Yalan Hu Endocrine Laboratory, Department of Laboratory Medicine, University of Amsterdam, Amsterdam, The Netherlands
Amsterdam Gastroenterology, Endocrinology & Metabolism Research Institute, Amsterdam, The Netherlands

Search for other papers by Yalan Hu in
Google Scholar
PubMed
Close
,
Gemma F Codner The Mary Lyon Centre at MRC Harwell, Harwell Campus, Oxfordshire, UK

Search for other papers by Gemma F Codner in
Google Scholar
PubMed
Close
,
Michelle Stewart The Mary Lyon Centre at MRC Harwell, Harwell Campus, Oxfordshire, UK

Search for other papers by Michelle Stewart in
Google Scholar
PubMed
Close
,
Susanne E La Fleur Endocrine Laboratory, Department of Laboratory Medicine, University of Amsterdam, Amsterdam, The Netherlands
Amsterdam Gastroenterology, Endocrinology & Metabolism Research Institute, Amsterdam, The Netherlands
Amsterdam Neuroscience, Cellular and Molecular Mechanisms, Amsterdam, The Netherlands

Search for other papers by Susanne E La Fleur in
Google Scholar
PubMed
Close
,
Paul A S van Trotsenburg Amsterdam Gastroenterology, Endocrinology & Metabolism Research Institute, Amsterdam, The Netherlands
Department of Pediatric Endocrinology, Emma Children’s Hospital, Amsterdam, The Netherlands

Search for other papers by Paul A S van Trotsenburg in
Google Scholar
PubMed
Close
,
Eric Fliers Amsterdam Gastroenterology, Endocrinology & Metabolism Research Institute, Amsterdam, The Netherlands
Department of Endocrinology, University of Amsterdam, Amsterdam, The Netherlands

Search for other papers by Eric Fliers in
Google Scholar
PubMed
Close
,
Raoul C Hennekam Department of Pediatrics, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands

Search for other papers by Raoul C Hennekam in
Google Scholar
PubMed
Close
, and
Anita Boelen Endocrine Laboratory, Department of Laboratory Medicine, University of Amsterdam, Amsterdam, The Netherlands
Amsterdam Gastroenterology, Endocrinology & Metabolism Research Institute, Amsterdam, The Netherlands
Amsterdam Reproduction & Development Research Institute, Amsterdam, The Netherlands

Search for other papers by Anita Boelen in
Google Scholar
PubMed
Close

Humans with the mutation Y509C in transducin beta like 1 X-linked (TBL1X HGNC ID HGNC:11585) have been reported to present with the combination of central congenital hypothyroidism and impaired hearing. TBL1X belongs to the WD40 repeat-containing protein family, is part of NCoR and SMRT corepressor complexes, and thereby involved in thyroid hormone signaling. In order to investigate the effects of the Y509C mutation in TBL1X on cellular thyroid hormone action, we aimed to generate a hemizygous male mouse cohort carrying the Tbl1x Y459C mutation which is equivalent to the human TBL1X Y509C mutation using CRISPR/Cas9 technology. Hemizygous male mice were small at birth and inactive. Their life span (median life span 93 days) was very short compared with heterozygous female mice (survived to >200 days with no welfare issues). About 52% of mice did not survive to weaning (133 mice). Of the remaining 118 mice, only 8 were hemizygous males who were unable to mate whereby it was impossible to generate homozygous female mice. In conclusion, the Tbl1x Y459C mutation in male mice has a marked negative effect on birth weight, survival, and fertility of male mice. The present findings are unexpected as they are in contrast to the mild phenotype in human males carrying the equivalent TBL1X Y509C mutation.

Restricted access
Weiye Zhao Department of Biology, University of York, York, UK

Search for other papers by Weiye Zhao in
Google Scholar
PubMed
Close
,
Susanna F Rose Department of Biology, University of York, York, UK

Search for other papers by Susanna F Rose in
Google Scholar
PubMed
Close
,
Ryan Blake CRUK Cambridge Institute, University of Cambridge, Cambridge, UK

Search for other papers by Ryan Blake in
Google Scholar
PubMed
Close
,
Aňze Godicelj Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Smith Building, Boston, Massachusetts, USA

Search for other papers by Aňze Godicelj in
Google Scholar
PubMed
Close
,
Amy E Cullen CRUK Cambridge Institute, University of Cambridge, Cambridge, UK

Search for other papers by Amy E Cullen in
Google Scholar
PubMed
Close
,
Jack Stenning Department of Biology, University of York, York, UK

Search for other papers by Jack Stenning in
Google Scholar
PubMed
Close
,
Lucy Beevors The Institute of Metabolism and Systems Research (IMSR), University of Birmingham, College of Medical and Dental Sciences, Birmingham, UK

Search for other papers by Lucy Beevors in
Google Scholar
PubMed
Close
,
Marcel Gehrung CRUK Cambridge Institute, University of Cambridge, Cambridge, UK

Search for other papers by Marcel Gehrung in
Google Scholar
PubMed
Close
,
Sanjeev Kumar Chris O’Brien Lifehouse, Sydney, New South Wales, Australia

Search for other papers by Sanjeev Kumar in
Google Scholar
PubMed
Close
,
Kamal Kishore CRUK Cambridge Institute, University of Cambridge, Cambridge, UK

Search for other papers by Kamal Kishore in
Google Scholar
PubMed
Close
,
Ashley Sawle CRUK Cambridge Institute, University of Cambridge, Cambridge, UK

Search for other papers by Ashley Sawle in
Google Scholar
PubMed
Close
,
Matthew Eldridge CRUK Cambridge Institute, University of Cambridge, Cambridge, UK

Search for other papers by Matthew Eldridge in
Google Scholar
PubMed
Close
,
Federico M Giorgi Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy

Search for other papers by Federico M Giorgi in
Google Scholar
PubMed
Close
,
Katherine S Bridge Department of Biology, University of York, York, UK
York Biomedical Research Institute, University of York, York, UK

Search for other papers by Katherine S Bridge in
Google Scholar
PubMed
Close
,
Florian Markowetz CRUK Cambridge Institute, University of Cambridge, Cambridge, UK

Search for other papers by Florian Markowetz in
Google Scholar
PubMed
Close
, and
Andrew N Holding Department of Biology, University of York, York, UK
York Biomedical Research Institute, University of York, York, UK
The Alan Turing Institute, Kings Cross, London, UK

Search for other papers by Andrew N Holding in
Google Scholar
PubMed
Close

The estrogen receptor-α (ER) drives 75% of breast cancers. On activation, the ER recruits and assembles a 1–2 MDa transcriptionally active complex. These complexes can modulate tumour growth, and understanding the roles of individual proteins within these complexes can help identify new therapeutic targets. Here, we present the discovery of ER and ZMIZ1 within the same multi-protein assembly by quantitative proteomics, and validated by proximity ligation assay. We characterise ZMIZ1 function by demonstrating a significant decrease in the proliferation of ER-positive cancer cell lines. To establish a role for the ER-ZMIZ1 interaction, we measured the transcriptional changes in the estrogen response post-ZMIZ1 knockdown using an RNA-seq time-course over 24 h. Gene set enrichment analysis of the ZMIZ1-knockdown data identified a specific delay in the response of estradiol-induced cell cycle genes. Integration of ENCODE data with our RNA-seq results identified that ER and ZMIZ1 both bind the promoter of E2F2. We therefore propose that ER and ZMIZ1 interact to enable the efficient estrogenic response at subset of cell cycle genes via a novel ZMIZ1–ER–E2F2 signalling axis. Finally, we show that high ZMIZ1 expression is predictive of worse patient outcome, ER and ZMIZ1 are co-expressed in breast cancer patients in TCGA and METABRIC, and the proteins are co-localised within the nuclei of tumour cell in patient biopsies. In conclusion, we establish that ZMIZ1 is a regulator of the estrogenic cell cycle response and provide evidence of the biological importance of the ER–ZMIZ1 interaction in ER-positive patient tumours, supporting potential clinical relevance.

Open access
Marta Santos-Hernández Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, UK

Search for other papers by Marta Santos-Hernández in
Google Scholar
PubMed
Close
,
Frank Reimann Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, UK

Search for other papers by Frank Reimann in
Google Scholar
PubMed
Close
, and
Fiona M Gribble Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, UK

Search for other papers by Fiona M Gribble in
Google Scholar
PubMed
Close

Enteroendocrine cells located along the gastrointestinal epithelium sense different nutrients/luminal contents that trigger the secretion of a variety of gut hormones with different roles in glucose homeostasis and appetite regulation. The incretin hormones glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) are involved in the regulation of insulin secretion, appetite, food intake and body weight after their nutrient-induced secretion from the gut. GLP-1 mimetics have been developed and used in the treatment of type 2 diabetes mellitus and obesity. Modulating the release of endogenous intestinal hormones may be a promising approach for the treatment of obesity and type 2 diabetes without surgery. For that reason, current understanding of the cellular mechanisms underlying intestinal hormone secretion will be the focus of this review. The mechanisms controlling hormone secretion depend on the nature of the stimulus, involving a variety of signalling pathways including ion channels, nutrient transporters and G-protein-coupled receptors.

Open access
Nirvay Sah Department of Pathology, University of California San Diego, La Jolla, California, USA
Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California, USA

Search for other papers by Nirvay Sah in
Google Scholar
PubMed
Close
and
Francesca Soncin Department of Pathology, University of California San Diego, La Jolla, California, USA
Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California, USA

Search for other papers by Francesca Soncin in
Google Scholar
PubMed
Close

Trophoblast stem cells (TSCs) are a proliferative multipotent population derived from the trophectoderm of the blastocyst, which will give rise to all the functional cell types of the trophoblast compartment of the placenta. The isolation and culture of TSCs in vitro represent a robust model to study mechanisms of trophoblast differentiation into mature cells both in successful and diseased pregnancy. Despite the highly conserved functions of the placenta, there is extreme variability in placental morphology, fetal–maternal interface, and development among eutherian mammals. This review aims to summarize the establishment and maintenance of TSCs in mammals such as primates, including human, rodents, and nontraditional animal models with a primary emphasis on epigenetic regulation of their origin while defining gaps in the current literature and areas of further development. FGF signaling is critical for mouse TSCs but dispensable for derivation of TSCs in other species. Human, simian, and bovine TSCs have much more complicated requirements of signaling pathways including activation of WNT and inhibition of TGFβ cascades. Epigenetic features such as DNA and histone methylation as well as histone acetylation are dynamic during development and are expressed in cell- and gestational age-specific pattern in placental trophoblasts. While TSCs from different species seem to recapitulate some select epigenomic features, there is a limitation in the comprehensive understanding of TSCs and how well TSCs retain placental epigenetic marks. Therefore, future studies should be directed at investigating epigenomic features of global and placental-specific gene expression in primary trophoblasts and TSCs.

Restricted access
Yan Meng School of Life Sciences, University of Nottingham, Queen’s Medical Centre, Nottingham, UK

Search for other papers by Yan Meng in
Google Scholar
PubMed
Close
,
Maria Toledo-Rodriguez School of Life Sciences, University of Nottingham, Queen’s Medical Centre, Nottingham, UK

Search for other papers by Maria Toledo-Rodriguez in
Google Scholar
PubMed
Close
,
Olena Fedorenko School of Life Sciences, University of Nottingham, Queen’s Medical Centre, Nottingham, UK

Search for other papers by Olena Fedorenko in
Google Scholar
PubMed
Close
, and
Paul A Smith School of Life Sciences, University of Nottingham, Queen’s Medical Centre, Nottingham, UK

Search for other papers by Paul A Smith in
Google Scholar
PubMed
Close

White adipose tissue (WAT) requires extracellular Ca2+ influx for lipolysis, differentiation, and expansion. This partly occurs via plasma membrane Ca2+ voltage-dependent channels (CaVs). However, WFA exists in different depots whose function varies with age, sex, and location. To explore whether their CaV expression profiles also differ we used RNAseq and qPCR on gonadal, mesenteric, retroperitoneal, and inguinal subcutaneous fat depots from rats of different ages and sex. CaV expression was found dependent on age, sex, and WFA location. In the gonadal depots of both sexes a significantly lower expression of CaV1.2 and CaV1.3 was seen for adults compared to pre-pubescent juveniles. A lower level of expression was also seen for CaV3.1 in adult male but not female gonadal WFA, the latter of whose expression remained unchanged with age. Relatively little expression of CaV3.2 and 3.2 was observed. In post-pubescent inguinal subcutaneous fat, where the third and fourth mammary glands are located, CaV3.1 was decreased in males but increased in females – thus suggesting that this channel is associated with mammogenesis; however, no difference in intracellular Ca2+ levels or adipocyte size were noted. For all adult depots, CaV3.1 expression was larger in females than males – a difference not seen in pre-pubescent rats. These observations are consistent with the changes of CaV3.1 expression seen in 3T3-L1 cell differentiation and the ability of selective CaV3.1 antagonists to inhibit adipogensis. Our results show that changes in CaV expression patterns occur in fat depots related to sexual dimorphism: reproductive tracts and mammogenesis.

Open access
Jenica H Kakadia Department of Biochemistry, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
Children's Health Research Institute, London, Ontario, Canada

Search for other papers by Jenica H Kakadia in
Google Scholar
PubMed
Close
,
Muhammad U Khalid Department of Biochemistry, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada

Search for other papers by Muhammad U Khalid in
Google Scholar
PubMed
Close
,
Ilka U Heinemann Department of Biochemistry, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada

Search for other papers by Ilka U Heinemann in
Google Scholar
PubMed
Close
, and
Victor K Han Department of Biochemistry, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
Children's Health Research Institute, London, Ontario, Canada
Department of Pediatrics, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada

Search for other papers by Victor K Han in
Google Scholar
PubMed
Close

Mechanisms underlying limitations in glucose supply that restrict fetal growth are not well established. IGF-1 is an important regulator of fetal growth and IGF-1 bioavailability is markedly inhibited by IGFBP-1 especially when the binding protein is hyperphosphorylated. We hypothesized that the AMPK–mTORC1 pathway increases IGFBP-1 phosphorylation in response to glucose deprivation. Glucose deprivation in HepG2 cells activated AMPK and TSC2, inhibited mTORC1 and increased IGFBP-1 secretion and site-specific phosphorylation. Glucose deprivation also decreased IGF-1 bioavailability and IGF-dependent activation of IGF-1R. AICAR (an AMPK activator) activated TSC2, inhibited mTORC1, and increased IGFBP-1 secretion/phosphorylation. Further, siRNA silencing of either AMPK or TSC2 prevented mTORC1 inhibition and IGFBP-1 secretion and phosphorylation in glucose deprivation. Our data suggest that the increase in IGFBP-1 phosphorylation in response to glucose deprivation is mediated by the activation of AMPK/TSC2 and inhibition of mTORC1, providing a possible mechanistic link between glucose deprivation and restricted fetal growth.

Open access
Xiaosa Li Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China

Search for other papers by Xiaosa Li in
Google Scholar
PubMed
Close
,
Chao Fan Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China

Search for other papers by Chao Fan in
Google Scholar
PubMed
Close
,
Jiale Wang Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China

Search for other papers by Jiale Wang in
Google Scholar
PubMed
Close
,
Ping Li Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China

Search for other papers by Ping Li in
Google Scholar
PubMed
Close
,
Xingyan Xu Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China

Search for other papers by Xingyan Xu in
Google Scholar
PubMed
Close
,
Ruixin Guo Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China

Search for other papers by Ruixin Guo in
Google Scholar
PubMed
Close
,
Jinzhi Wei Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China

Search for other papers by Jinzhi Wei in
Google Scholar
PubMed
Close
,
Yang Cheng Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, China

Search for other papers by Yang Cheng in
Google Scholar
PubMed
Close
,
Huiping Lin Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China

Search for other papers by Huiping Lin in
Google Scholar
PubMed
Close
, and
Xiaodong Fu Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
Qingyuan People’s Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, Guangdong, China

Search for other papers by Xiaodong Fu in
Google Scholar
PubMed
Close

Follicle-stimulating hormone (FSH) accelerates osteoporosis in postmenopausal women, while the underlying mechanism remains uncharacterized. N6-methyladenosine (m6A) is one of the most important regulations in the development of osteoporosis. In this study, we aimed to investigate the role of FSH in m6A modification and osteoclast function. Here, we showed that FSH upregulated m6A levels in osteoclasts via stimulating methyltransferase-like 3 (METTL3) protein expression. FSH enhanced osteoclast migration, while the knockdown of METTL3 eliminated this enhancement. Both MeRIP-seq and RNA sequencing identified that cathepsin K (CTSK) is the potential downstream target of METTL3. Knockdown of CTSK reduced FSH-upregulated osteoclast migration. Furthermore, silencing METTL3 decreased CTSK mRNA stability. Finally, FSH induced phosphorylation of cyclic-AMP response element-binding protein (CREB), while silencing of CREB attenuated the effects of FSH on the promoter transcriptional activity of Mettl3 and CTSK/METTL3 protein. Taken together, these findings indicate that FSH promotes osteoclast migration via the CREB/METTL3/CTSK signaling pathway, which may provide a potential target for suppressing osteoclast mobility and postmenopausal osteoporosis therapy.

Restricted access
Isabelle Durrer Department of Nephrology and Hypertension University of Bern, Berne, Switzerland

Search for other papers by Isabelle Durrer in
Google Scholar
PubMed
Close
,
Daniel Ackermann Department of Nephrology and Hypertension University of Bern, Berne, Switzerland

Search for other papers by Daniel Ackermann in
Google Scholar
PubMed
Close
,
Rahel Klossner Department of Nephrology and Hypertension University of Bern, Berne, Switzerland
Department of Internal Medicine, Sonnenhof, Lindenhofgruppe, Berne, Switzerland

Search for other papers by Rahel Klossner in
Google Scholar
PubMed
Close
,
Michael Grössl Department of Nephrology and Hypertension University of Bern, Berne, Switzerland

Search for other papers by Michael Grössl in
Google Scholar
PubMed
Close
,
Clarissa Vögel Department of Nephrology and Hypertension University of Bern, Berne, Switzerland

Search for other papers by Clarissa Vögel in
Google Scholar
PubMed
Close
,
Therina Du Toit Department for BioMedical Research University of Bern, Berne, Switzerland

Search for other papers by Therina Du Toit in
Google Scholar
PubMed
Close
,
Bruno Vogt Department of Nephrology and Hypertension University of Bern, Berne, Switzerland

Search for other papers by Bruno Vogt in
Google Scholar
PubMed
Close
,
Heidi Jamin Department of Nephrology and Hypertension University of Bern, Berne, Switzerland
Department for BioMedical Research University of Bern, Berne, Switzerland

Search for other papers by Heidi Jamin in
Google Scholar
PubMed
Close
,
Markus G Mohaupt Department of Internal Medicine, Sonnenhof, Lindenhofgruppe, Berne, Switzerland
Department for BioMedical Research University of Bern, Berne, Switzerland

Search for other papers by Markus G Mohaupt in
Google Scholar
PubMed
Close
, and
Carine Gennari-Moser Department of Nephrology and Hypertension University of Bern, Berne, Switzerland
Department for BioMedical Research University of Bern, Berne, Switzerland

Search for other papers by Carine Gennari-Moser in
Google Scholar
PubMed
Close

Extra-adrenal de novo aldosterone (Aldo) production has been described inconsistently. Systematic data based upon state-of-the-art technology including validated controls are sparse. We hypothesized that aldosterone synthase (CYP11B2) expression and de novo Aldo production are absent in nonadrenal human cell lines, either immortalized cell lines or commercially available primary cell lines, including peripheral blood mononuclear cells (PBMCs) of individuals without and with primary hyperaldosteronism (PA). CYP11B2-transfected COS-7 and endogenous CYP11B2 expressing adrenal H295R cells served as positive controls. Various well-characterized, purchased, immortalized (BeWo, HEK293, HTR-8/SVneo, JEG-3) and primary (HAEC, HLEC, HRGEC, HRMC, HUAEC, HUVEC, PBMC) cell lines as well as self-isolated PBMCs from PA patients (n = 5) were incubated with the steroid hormone substrates progesterone, deoxycorticosterone, corticosterone or 18-OH-corticosterone with and without Ang II for 24 h to assess CYP11B2 enzymatic activity. CYP11B2 expression was analyzed by real-time PCR and liquid chromatography–mass spectrometry was used to quantify Aldo production. Pronounced CYP11B2 mRNA expression and Aldo production were observed in both positive controls, which followed an incremental time course. Neither substrates alone nor coincubation with Ang II significantly stimulated CYP11B2 expression or Aldo production in various immortalized and primary cell lines and PBMCs of PA patients. These results strongly support the absence of relevant de novo extra-adrenal Aldo production in nonadrenal cells, including blood mononuclear cells, irrespective of the absence or presence of autonomous adrenal Aldo production.

Open access