Fibroblast growth factor 23 (Fgf23) is produced by bone and functions primarily as a phosphaturia hormone. We previously reported that overexpression of the Notch intracellular domain (NICD) in osteogenic cells enhances Fgf23 expression in association with osteomalacia in vivo. Here, we investigated the underlying mechanisms using osteogenic cell lines UMR-106 and IDG-SW3 cells. NICD overexpression increased Fgf23 levels in both cell types. Manipulating RBPJ-κ activity, either a dominant-negative or constitutively active form, revealed that Notch-mediated Fgf23 expression is dependent on RBPJ-κ. Treatment with iron chelator Desferrioxamine (DFO) upregulated Fgf23 expression, which was abolished by dominant-negative RBPJ-κ overexpression. This effect was partially attenuated by short hairpin RNA (shRNA) targeting hypoxia-inducible factor (HIF)-2α, but not HIF-1α. DFO treatment also increased expression of Notch1 protein, but not Notch2 and Nocth3, in parallel with upregulation of the Notch target mRNAs, Hes1 and Hey1. In addition, DFO elevated the expression of γ-secretase subunits, whereas a γ-secretase inhibitor suppressed DFO-induced increases in Notch1 and Fgf23 levels, suggesting that increased γ-secretase expression promotes Notch processing. Moreover, Notch signaling exerted an additive stimulatory effect on parathyroid hormone (PTH)-induced Fgf23 expression, at least in part through interaction with the protein kinase A (PKA) pathway. Co-immunoprecipitation assays revealed a physical interaction between NICD and CREB period Collectively, these findings demonstrate that Notch signaling regulates Fgf23 expression through crosstalk with hypoxic and PTH pathways, providing novel insights into Fgf23 regulation and identifying potential therapeutic targets for Fgf23-related disorders.
{"title":"Notch signaling modulates Fgf23 expression through crosstalk with hypoxia and PTH pathways in osteogenic cells","authors":"Yoshihiro Tamamura , Kenta Terai , Akira Yamaguchi","doi":"10.1016/j.mce.2025.112663","DOIUrl":"10.1016/j.mce.2025.112663","url":null,"abstract":"<div><div>Fibroblast growth factor 23 (Fgf23) is produced by bone and functions primarily as a phosphaturia hormone. We previously reported that overexpression of the Notch intracellular domain (NICD) in osteogenic cells enhances Fgf23 expression in association with osteomalacia <em>in vivo</em>. Here, we investigated the underlying mechanisms using osteogenic cell lines UMR-106 and IDG-SW3 cells. NICD overexpression increased <em>Fgf23</em> levels in both cell types. Manipulating RBPJ-κ activity, either a dominant-negative or constitutively active form, revealed that Notch-mediated <em>Fgf23</em> expression is dependent on RBPJ-κ. Treatment with iron chelator Desferrioxamine (DFO) upregulated <em>Fgf23</em> expression, which was abolished by dominant-negative RBPJ-κ overexpression. This effect was partially attenuated by short hairpin RNA (shRNA) targeting hypoxia-inducible factor (<em>HIF</em>)-2α, but not <em>HIF-1</em>α. DFO treatment also increased expression of Notch1 protein, but not Notch2 and Nocth3, in parallel with upregulation of the Notch target mRNAs, <em>Hes1</em> and <em>Hey1</em>. In addition, DFO elevated the expression of γ<strong><em>-</em></strong>secretase subunits, whereas a γ<strong>-</strong>secretase inhibitor suppressed DFO-induced increases in Notch1 and Fgf23 levels, suggesting that increased γ<strong><em>-</em></strong>secretase expression promotes Notch processing. Moreover, Notch signaling exerted an additive stimulatory effect on parathyroid hormone (PTH)-induced <em>Fgf23</em> expression, at least in part through interaction with the protein kinase A (PKA) pathway. Co-immunoprecipitation assays revealed a physical interaction between NICD and CREB period Collectively, these findings demonstrate that Notch signaling regulates Fgf23 expression through crosstalk with hypoxic and PTH pathways, providing novel insights into Fgf23 regulation and identifying potential therapeutic targets for Fgf23-related disorders.</div></div>","PeriodicalId":18707,"journal":{"name":"Molecular and Cellular Endocrinology","volume":"610 ","pages":"Article 112663"},"PeriodicalIF":3.6,"publicationDate":"2025-09-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145138251","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-09-19DOI: 10.1016/j.mce.2025.112662
María Laura Leonardi , Alejandro Ranieri , Cintia Romina Gatti , Evangelina Capobianco , Alicia Jawerbaum , Romina Higa
Introduction
Advanced maternal age (AMA) is associated with increased risks of adverse pregnancy outcomes partly due to placental dysfunction; however, the underlying mechanisms remain poorly understood. This study aimed to investigate early placental development in AMA pregnancies, focusing on FoxO1 activation and its role in cellular senescence and oxidative stress.
Methods
Three-month-old (Control) and 10-month-old (AMA) Wistar rats were mated with young males. On day 12 of pregnancy, FOXO1 activity and the expression of its target genes, oxidative status and morphometry were evaluated in the decidua and developing placenta.
Results
AMA rats exhibited a reduced number of implantation sites, fewer viable embryos, and decreased embryonic crown-rump length, indicating restricted growth. Markers of oxidative stress were increased in the decidua. At the molecular level, FOXO1 phosphorylation was reduced in the decidua, suggesting increased FOXO1 activation, whereas in the developing placenta, FOXO1 phosphorylation was elevated, indicating its inactivation. SGK1, a kinase that regulates FOXO1 phosphorylation, showed decreased phosphorylation in the decidua of AMA rats. Moreover, the senescence markers Cdkn1a (P21) and Cdkn2a (P16), known FOXO1 target genes, were upregulated in the decidua and downregulated in the developing placenta. These changes were associated with impaired cell proliferation in the decidua and a reduced syncitiotrophoblast layer in the developing placenta.
Conclusion
These findings highlight the differential regulation of FOXO1 in the decidua and placenta during AMA pregnancies. Increased FOXO1 activity in the decidua, likely driven by oxidative stress, and reduced SGK1 phosphorylation, may impair decidual function and contribute to altered placenta development with reduced FOXO1 activity.
{"title":"Advanced maternal age induced altered FoxO1 activation and cellular senescence in early placenta development in rats","authors":"María Laura Leonardi , Alejandro Ranieri , Cintia Romina Gatti , Evangelina Capobianco , Alicia Jawerbaum , Romina Higa","doi":"10.1016/j.mce.2025.112662","DOIUrl":"10.1016/j.mce.2025.112662","url":null,"abstract":"<div><h3>Introduction</h3><div>Advanced maternal age (AMA) is associated with increased risks of adverse pregnancy outcomes partly due to placental dysfunction; however, the underlying mechanisms remain poorly understood. This study aimed to investigate early placental development in AMA pregnancies, focusing on FoxO1 activation and its role in cellular senescence and oxidative stress.</div></div><div><h3>Methods</h3><div>Three-month-old (Control) and 10-month-old (AMA) Wistar rats were mated with young males. On day 12 of pregnancy, FOXO1 activity and the expression of its target genes, oxidative status and morphometry were evaluated in the decidua and developing placenta.</div></div><div><h3>Results</h3><div>AMA rats exhibited a reduced number of implantation sites, fewer viable embryos, and decreased embryonic crown-rump length, indicating restricted growth. Markers of oxidative stress were increased in the decidua. At the molecular level, FOXO1 phosphorylation was reduced in the decidua, suggesting increased FOXO1 activation, whereas in the developing placenta, FOXO1 phosphorylation was elevated, indicating its inactivation. SGK1, a kinase that regulates FOXO1 phosphorylation, showed decreased phosphorylation in the decidua of AMA rats. Moreover, the senescence markers <em>Cdkn1a</em> (P21) and <em>Cdkn2a</em> (P16), known FOXO1 target genes, were upregulated in the decidua and downregulated in the developing placenta. These changes were associated with impaired cell proliferation in the decidua and a reduced syncitiotrophoblast layer in the developing placenta.</div></div><div><h3>Conclusion</h3><div>These findings highlight the differential regulation of FOXO1 in the decidua and placenta during AMA pregnancies. Increased FOXO1 activity in the decidua, likely driven by oxidative stress, and reduced SGK1 phosphorylation, may impair decidual function and contribute to altered placenta development with reduced FOXO1 activity.</div></div>","PeriodicalId":18707,"journal":{"name":"Molecular and Cellular Endocrinology","volume":"610 ","pages":"Article 112662"},"PeriodicalIF":3.6,"publicationDate":"2025-09-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145113819","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-09-18DOI: 10.1016/j.mce.2025.112661
Di Wu , Kejia Zhang , Jiachen Tan , Faheem Ahmed Khan , Chunjie Huang , Fei Sun
Efferocytosis is energy-consuming, and continuous efferocytosis imposes metabolic burdens on the phagocytes. Sertoli cells (SCs) are specialized phagocytes in the testis for efferocytosis of non-viable germ cells and residual bodies. What remains elusive is how SCs integrate metabolic adaptations in response to efferocytosis. Here, we identify the Nrf2/Bach1 axis as an important molecular machinery of SC-mediated efferocytosis. Nrf2 activation during efferocytosis stabilizes Bach1 expression. Nrf2 activation or Bach1 overexpression promotes SC-mediated efferocytosis, while the opposite phenotype is incurred by Nrf2 inactivation or Bach1 deficiency, with oxidative stress being a contributing factor. Beyond experiencing attenuated glucose uptake and ATP production, Bach1-deficient SCs exhibit a reduced NAD+/NADH ratio, and restraining NAD+ consumption by inhibiting serine biosynthesis rescues their impaired efferocytosis. We further observe an up-regulation of anti-ferroptotic genes in SCs upon Bach1 deficiency and demonstrate a protective role of ferroptosis in this scenario. We thus propose that redox homeostasis and energy metabolism lie at the nexus of the Nrf2/Bach1 axis in the regulation of SC-mediated efferocytosis. Our study explores the regulatory role of the Nrf2/Bach1 axis in SC-mediated efferocytosis, which will lead to a better appreciation of SCs in male reproductive health.
{"title":"Nrf2/Bach1 axis regulates redox homeostasis and energy metabolism to optimize Sertoli cell-mediated efferocytosis","authors":"Di Wu , Kejia Zhang , Jiachen Tan , Faheem Ahmed Khan , Chunjie Huang , Fei Sun","doi":"10.1016/j.mce.2025.112661","DOIUrl":"10.1016/j.mce.2025.112661","url":null,"abstract":"<div><div>Efferocytosis is energy-consuming, and continuous efferocytosis imposes metabolic burdens on the phagocytes. Sertoli cells (SCs) are specialized phagocytes in the testis for efferocytosis of non-viable germ cells and residual bodies. What remains elusive is how SCs integrate metabolic adaptations in response to efferocytosis. Here, we identify the Nrf2/Bach1 axis as an important molecular machinery of SC-mediated efferocytosis. Nrf2 activation during efferocytosis stabilizes Bach1 expression. Nrf2 activation or Bach1 overexpression promotes SC-mediated efferocytosis, while the opposite phenotype is incurred by Nrf2 inactivation or Bach1 deficiency, with oxidative stress being a contributing factor. Beyond experiencing attenuated glucose uptake and ATP production, Bach1-deficient SCs exhibit a reduced NAD<sup>+</sup>/NADH ratio, and restraining NAD<sup>+</sup> consumption by inhibiting serine biosynthesis rescues their impaired efferocytosis. We further observe an up-regulation of anti-ferroptotic genes in SCs upon Bach1 deficiency and demonstrate a protective role of ferroptosis in this scenario. We thus propose that redox homeostasis and energy metabolism lie at the nexus of the Nrf2/Bach1 axis in the regulation of SC-mediated efferocytosis. Our study explores the regulatory role of the Nrf2/Bach1 axis in SC-mediated efferocytosis, which will lead to a better appreciation of SCs in male reproductive health.</div></div>","PeriodicalId":18707,"journal":{"name":"Molecular and Cellular Endocrinology","volume":"610 ","pages":"Article 112661"},"PeriodicalIF":3.6,"publicationDate":"2025-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145102812","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-09-16DOI: 10.1016/j.mce.2025.112660
Xiaoyu Liu , Guoyan Xu , Yunlu Xu , Yuling Xu
Background
Atherosclerosis (AS) is a chronic vascular disease, and perivascular adipose tissue dysfunction is an important cause of the arterial plaque formation involved. However, the underlying mechanism has not been fully elucidated. The aim of this study was to investigate the mechanism of oxidized low-density lipoprotein (ox-LDL) stimulation of macrophage-derived exosomes in the development of AS.
Methods
We isolated exosomes from ox-LDL-treated macrophages and injected them into Western diet-fed ApoE−/− mice. We assessed AS, lipid metabolism, and endothelial function by histology, ELISA, qPCR, and western blotting, and examined BMP7 and OPA1 regulation in brown fat and vascular endothelium.
Results
Macrophage-derived exosomes were extracted, and their size was determined by transmission electron microscopy. Additionally, CD9, CD63, and TSG101 protein expression within these macrophages was determined. Compared with the control group, the exosomes group showed increased expression of AP2 and PPAR and decreased expression of UCP-1, PGC-1α, and BMP7. Furthermore, when BMP7 was knocked down, the expression of the lipid metabolites FASN, SCD1, HSL, and ATGL as well as of OPA1 decreased. In an ApoE−/− mouse model, compared to the control group, increased arterial plaques and plaque lesion formation were observed in the exosome group, along with elevated expression of the lipid metrics TC, TG, LDL-C, and HDL-C and significant increases in the expression of the proinflammatory factors VCAM1, ICAM1, MCP-1, and IL-6. Consequently the progression of AS was aggravated in this group.
Conclusions
This study demonstrated that ox-LDL stimulated exosome secretion from macrophages, accelerating the AS process. It also showed that, mechanistically, BMP7 regulates the expression of OPA1 and affects the normal lipid metabolism, thereby accelerating AS.
{"title":"Ox-LDL-stimulated macrophage-derived exosomes regulate adipose tissue remodeling and promote the progression of atherosclerosis","authors":"Xiaoyu Liu , Guoyan Xu , Yunlu Xu , Yuling Xu","doi":"10.1016/j.mce.2025.112660","DOIUrl":"10.1016/j.mce.2025.112660","url":null,"abstract":"<div><h3>Background</h3><div>Atherosclerosis (AS) is a chronic vascular disease, and perivascular adipose tissue dysfunction is an important cause of the arterial plaque formation involved. However, the underlying mechanism has not been fully elucidated. The aim of this study was to investigate the mechanism of oxidized low-density lipoprotein (ox-LDL) stimulation of macrophage-derived exosomes in the development of AS.</div></div><div><h3>Methods</h3><div>We isolated exosomes from ox-LDL-treated macrophages and injected them into Western diet-fed ApoE<sup>−/−</sup> mice. We assessed AS, lipid metabolism, and endothelial function by histology, ELISA, qPCR, and western blotting, and examined BMP7 and OPA1 regulation in brown fat and vascular endothelium.</div></div><div><h3>Results</h3><div>Macrophage-derived exosomes were extracted, and their size was determined by transmission electron microscopy. Additionally, CD9, CD63, and TSG101 protein expression within these macrophages was determined. Compared with the control group, the exosomes group showed increased expression of AP2 and PPAR and decreased expression of UCP-1, PGC-1α, and BMP7. Furthermore, when BMP7 was knocked down, the expression of the lipid metabolites FASN, SCD1, HSL, and ATGL as well as of OPA1 decreased. In an ApoE<sup>−/−</sup> mouse model, compared to the control group, increased arterial plaques and plaque lesion formation were observed in the exosome group, along with elevated expression of the lipid metrics TC, TG, LDL-C, and HDL-C and significant increases in the expression of the proinflammatory factors VCAM1, ICAM1, MCP-1, and IL-6. Consequently the progression of AS was aggravated in this group.</div></div><div><h3>Conclusions</h3><div>This study demonstrated that ox-LDL stimulated exosome secretion from macrophages, accelerating the AS process. It also showed that, mechanistically, BMP7 regulates the expression of OPA1 and affects the normal lipid metabolism, thereby accelerating AS.</div></div>","PeriodicalId":18707,"journal":{"name":"Molecular and Cellular Endocrinology","volume":"609 ","pages":"Article 112660"},"PeriodicalIF":3.6,"publicationDate":"2025-09-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145086380","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-09-16DOI: 10.1016/j.mce.2025.112659
Cigdem Aydin Acar , Suray Pehlivanoglu
This study synthesized silver nanoparticles using cherry stem aqueous extract (Cs-AgNPs) and evaluated their physicochemical properties, cytotoxic effects on pancreatic β-cell lines, antioxidant activity, and their potential to enhance insulin secretion in 3D pancreatic β-cell models. Cs-AgNPs were synthesized via a reaction between cherry stem extract and silver nitrate, confirmed through color change and UV–Vis spectrophotometry. Characterization using EDS, TEM, and XRD revealed spherical nanoparticles with a crystalline structure, sizes ranging from 10.93 to 31.18 nm, and an average size of 26.67 nm. Biological assessments showed dose-dependent cytotoxic effects on pancreatic β-cell lines, with reduced viability observed at ≥2 μg/mL for INS-1 cells and ≥5 μg/mL for RINm5F cells. Antioxidant activity was confirmed through ABTS assay, with an IC50 value of 78.81 μg/mL. Functional studies on 3D pancreatic β-cell spheroids revealed a significant 1.6-fold increase in insulin secretion in RINm5F cells (p = 0.0166) and a modest 1.2-fold increase in INS-1 cells. The results highlight the antioxidant properties and insulin secretion enhancement potential of Cs-AgNPs, suggesting their promise for diabetes-related applications. Further research is recommended to explore their therapeutic benefits and expand their biomedical utility.
{"title":"Three-dimensional (3D) pancreatic β-cell models reveal insulin-secretion enhancing potential of green synthesized silver nanoparticles","authors":"Cigdem Aydin Acar , Suray Pehlivanoglu","doi":"10.1016/j.mce.2025.112659","DOIUrl":"10.1016/j.mce.2025.112659","url":null,"abstract":"<div><div>This study synthesized silver nanoparticles using cherry stem aqueous extract (Cs-AgNPs) and evaluated their physicochemical properties, cytotoxic effects on pancreatic β-cell lines, antioxidant activity, and their potential to enhance insulin secretion in 3D pancreatic β-cell models. Cs-AgNPs were synthesized via a reaction between cherry stem extract and silver nitrate, confirmed through color change and UV–Vis spectrophotometry. Characterization using EDS, TEM, and XRD revealed spherical nanoparticles with a crystalline structure, sizes ranging from 10.93 to 31.18 nm, and an average size of 26.67 nm. Biological assessments showed dose-dependent cytotoxic effects on pancreatic β-cell lines, with reduced viability observed at ≥2 μg/mL for INS-1 cells and ≥5 μg/mL for RINm5F cells. Antioxidant activity was confirmed through ABTS assay, with an IC<sub>50</sub> value of 78.81 μg/mL. Functional studies on 3D pancreatic β-cell spheroids revealed a significant 1.6-fold increase in insulin secretion in RINm5F cells (p = 0.0166) and a modest 1.2-fold increase in INS-1 cells. The results highlight the antioxidant properties and insulin secretion enhancement potential of Cs-AgNPs, suggesting their promise for diabetes-related applications. Further research is recommended to explore their therapeutic benefits and expand their biomedical utility.</div></div>","PeriodicalId":18707,"journal":{"name":"Molecular and Cellular Endocrinology","volume":"609 ","pages":"Article 112659"},"PeriodicalIF":3.6,"publicationDate":"2025-09-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145086387","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-09-13DOI: 10.1016/j.mce.2025.112657
Daniel M. Gallagher , Md Zahidul Islam Khan , Steven Patterson , Finbarr P.M. O'Harte , Nigel Irwin
Objective
The neuropeptide spexin is recognised as a satiety-inducing hormone, but overall effects on metabolism are less characterised. Rapid enzymatic metabolism means elucidating biological effects of spexin is challenging, because the bioactive profile is short.
Methods
Therefore, in the present study, an Asp1 for Asn1 substituted spexin analogue, D1Spx, alongside two related fatty acid derivatised analogues, (γGlu-Pal)-D1Spx and (K11γGlu-Pal)-D1Spx, were synthesised and effects on pancreatic beta-cell secretory function and health investigated together with impact on appetite and glucose homeostasis in mice.
Results
Spexin immunoreactivity was initially confirmed in BRIN-BD11 beta-cells. Interestingly, like native spexin, D1Spx was liable to plasma enzyme degradation, but the fatty acid derivatised molecules remained intact. None of the peptides augmented insulin secretion from BRIN-BD11 cells. Moreover, the spexin peptides inhibited alanine‐induced insulin secretion, with native spexin having no effect on intracellular calcium. However, all spexin peptides (10−8 and 10−6 M) promoted beta-cell proliferation, whilst native spexin and (γGlu-Pal)-D1Spx protected against cytokine-induced beta-cell apoptosis. When administered intraperitoneally to mice, spexin peptides lacked effects on appetite regulation, even at elevated doses of 250 nmol/kg. Following conjoint injection with saline, none of the spexin peptides affected blood glucose levels barring a negligible increase by D1Spx. When administered together with glucose, (γGlu-Pal)-D1Spx slightly increased blood glucose at 30 min post-injection, but there was no overall difference between the spexin peptides when compared to glucose alone.
Conclusions
Acylation creates stable spexin analogues with similar bioactivity as native spexin, including promotion of beta-cell proliferation and partial protection against apoptosis.
{"title":"Impact of novel amino acid substituted and acylated spexin analogues on pancreatic beta-cell function, appetite and glucose homeostasis","authors":"Daniel M. Gallagher , Md Zahidul Islam Khan , Steven Patterson , Finbarr P.M. O'Harte , Nigel Irwin","doi":"10.1016/j.mce.2025.112657","DOIUrl":"10.1016/j.mce.2025.112657","url":null,"abstract":"<div><h3>Objective</h3><div>The neuropeptide spexin is recognised as a satiety-inducing hormone, but overall effects on metabolism are less characterised. Rapid enzymatic metabolism means elucidating biological effects of spexin is challenging, because the bioactive profile is short.</div></div><div><h3>Methods</h3><div>Therefore, in the present study, an Asp<sup>1</sup> for Asn<sup>1</sup> substituted spexin analogue, D<sup>1</sup>Spx, alongside two related fatty acid derivatised analogues, (γGlu-Pal)-D<sup>1</sup>Spx and (K<sup>11</sup>γGlu-Pal)-D<sup>1</sup>Spx, were synthesised and effects on pancreatic beta-cell secretory function and health investigated together with impact on appetite and glucose homeostasis in mice.</div></div><div><h3>Results</h3><div>Spexin immunoreactivity was initially confirmed in BRIN-BD11 beta-cells. Interestingly, like native spexin, D<sup>1</sup>Spx was liable to plasma enzyme degradation, but the fatty acid derivatised molecules remained intact. None of the peptides augmented insulin secretion from BRIN-BD11 cells. Moreover, the spexin peptides inhibited alanine‐induced insulin secretion, with native spexin having no effect on intracellular calcium. However, all spexin peptides (10<sup>−8</sup> and 10<sup>−6</sup> M) promoted beta-cell proliferation, whilst native spexin and (γGlu-Pal)-D<sup>1</sup>Spx protected against cytokine-induced beta-cell apoptosis. When administered intraperitoneally to mice, spexin peptides lacked effects on appetite regulation, even at elevated doses of 250 nmol/kg. Following conjoint injection with saline, none of the spexin peptides affected blood glucose levels barring a negligible increase by D<sup>1</sup>Spx. When administered together with glucose, (γGlu-Pal)-D<sup>1</sup>Spx slightly increased blood glucose at 30 min post-injection, but there was no overall difference between the spexin peptides when compared to glucose alone.</div></div><div><h3>Conclusions</h3><div>Acylation creates stable spexin analogues with similar bioactivity as native spexin, including promotion of beta-cell proliferation and partial protection against apoptosis.</div></div>","PeriodicalId":18707,"journal":{"name":"Molecular and Cellular Endocrinology","volume":"609 ","pages":"Article 112657"},"PeriodicalIF":3.6,"publicationDate":"2025-09-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145070028","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
<div><div>Over the past few decades, a significant change globally in sugar intake has coincided with a rising incidence of male infertility, which is now a major public health concern. Diets rich in fructose have been implicated in both male infertility and increased susceptibility to metabolic disorders, such as obesity, diabetes, and related heart problems. While fructose is known to be present in seminal fluid and crucial for sperm motility, the precise role of fructose in testicular function remains largely unknown.</div><div>GLUT5 is an exclusive fructose transporter essential for dietary fructose uptake in the intestine. It is also expressed mainly in germ and Leydig cells. We recently revealed that disrupting the <em>Glut5</em> gene in male mice impairs spermatogenesis and steroidogenesis. However, its specific role within Leydig cells remains unexplored. Therefore, we investigated its role by inhibiting GLUT5 in a murine Leydig cell line (mLTC-1) using a specific inhibitor of GLUT5, MSNBA, combined with a multi-omics approach.</div><div>Exposing mLTC-1 cells to MSNBA reduced the intracellular fructose content, limited cell proliferation, and enhanced progesterone and androgens production (Δ4-androstenedione and testosterone). The latter was associated with the upregulation of two genes and proteins involved in steroidogenesis, such as <em>Hsd3b</em> and steroidogenic acute regulatory protein (<em>StAR</em>). GLUT5 inhibition in mLTC-1 cells also modified lipid and carbohydrate metabolism. Lipidomic analysis showed decreased cholesterol esters and a shift in the ratio of polyunsaturated fatty acids (PUFAs) to monounsaturated fatty acids (MUFAs). These lipid changes correlated with alterations in the expression of mRNA-encoding enzymes involved in lipogenesis, such as ELOVL6. Metabolomics analysis showed a reduction in most glycolysis metabolites, except for pyruvate and lactate. However, pyruvate could conserve its level by a production through an amino acid pathway using the higher branched-chain amino acid content. Nevertheless, the activity of mitochondria measured by seahorse was not altered. The transcriptomic analysis performed by BRB-seq approach revealed an upregulation of several androgen-sensitive genes, such as <em>Akap5, Slc39a9,</em> an androgen receptor or lactate dehydrogenase A (<em>Ldha</em>), which produces lactate, and downregulation of several genes associated with the insulin pathway such as <em>Tsc2 or</em> the hexokinase <em>Hkdc1</em>.</div><div>In conclusion, GLUT5 supported fructose intake in the murine Leydig cell line mLTC-1, leading to a reduction in cell proliferation. The consequences of inhibition of GLUT5 led to an increase in fatty acids cell content, a perturbation in glycolysis and amino-acid metabolism but an enhanced androgen production. Since androgens regulate spermatogenesis, hyperandrogenism induced by a lower fructose content in Leydig cells may be a primary cause leading to the disruption of spe
{"title":"Inhibiting the fructose transporter GLUT5 boosts testosterone production in a murine mLTC-1 leydig cell line","authors":"Aikaterini Kallianioti , Guillaume Bourdon , Claire Chevaleyre , Christine Péchoux , Christelle Ramé , Jérôme Bourgeais , Olivier Hérault , Nancy Geoffre , Thomas Darde , Ingrid Plotton , Véronique Douard , Joëlle Dupont , Pascal Froment","doi":"10.1016/j.mce.2025.112658","DOIUrl":"10.1016/j.mce.2025.112658","url":null,"abstract":"<div><div>Over the past few decades, a significant change globally in sugar intake has coincided with a rising incidence of male infertility, which is now a major public health concern. Diets rich in fructose have been implicated in both male infertility and increased susceptibility to metabolic disorders, such as obesity, diabetes, and related heart problems. While fructose is known to be present in seminal fluid and crucial for sperm motility, the precise role of fructose in testicular function remains largely unknown.</div><div>GLUT5 is an exclusive fructose transporter essential for dietary fructose uptake in the intestine. It is also expressed mainly in germ and Leydig cells. We recently revealed that disrupting the <em>Glut5</em> gene in male mice impairs spermatogenesis and steroidogenesis. However, its specific role within Leydig cells remains unexplored. Therefore, we investigated its role by inhibiting GLUT5 in a murine Leydig cell line (mLTC-1) using a specific inhibitor of GLUT5, MSNBA, combined with a multi-omics approach.</div><div>Exposing mLTC-1 cells to MSNBA reduced the intracellular fructose content, limited cell proliferation, and enhanced progesterone and androgens production (Δ4-androstenedione and testosterone). The latter was associated with the upregulation of two genes and proteins involved in steroidogenesis, such as <em>Hsd3b</em> and steroidogenic acute regulatory protein (<em>StAR</em>). GLUT5 inhibition in mLTC-1 cells also modified lipid and carbohydrate metabolism. Lipidomic analysis showed decreased cholesterol esters and a shift in the ratio of polyunsaturated fatty acids (PUFAs) to monounsaturated fatty acids (MUFAs). These lipid changes correlated with alterations in the expression of mRNA-encoding enzymes involved in lipogenesis, such as ELOVL6. Metabolomics analysis showed a reduction in most glycolysis metabolites, except for pyruvate and lactate. However, pyruvate could conserve its level by a production through an amino acid pathway using the higher branched-chain amino acid content. Nevertheless, the activity of mitochondria measured by seahorse was not altered. The transcriptomic analysis performed by BRB-seq approach revealed an upregulation of several androgen-sensitive genes, such as <em>Akap5, Slc39a9,</em> an androgen receptor or lactate dehydrogenase A (<em>Ldha</em>), which produces lactate, and downregulation of several genes associated with the insulin pathway such as <em>Tsc2 or</em> the hexokinase <em>Hkdc1</em>.</div><div>In conclusion, GLUT5 supported fructose intake in the murine Leydig cell line mLTC-1, leading to a reduction in cell proliferation. The consequences of inhibition of GLUT5 led to an increase in fatty acids cell content, a perturbation in glycolysis and amino-acid metabolism but an enhanced androgen production. Since androgens regulate spermatogenesis, hyperandrogenism induced by a lower fructose content in Leydig cells may be a primary cause leading to the disruption of spe","PeriodicalId":18707,"journal":{"name":"Molecular and Cellular Endocrinology","volume":"610 ","pages":"Article 112658"},"PeriodicalIF":3.6,"publicationDate":"2025-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145065021","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-09-11DOI: 10.1016/j.mce.2025.112656
Pengtao Chen , Jinpeng Ruan , Fanzheng Xue , Xuejuan Dai , Chen Tang , Mingyue Chen , Nengming Xiao , Zhijian Cai , Chunyan Yang , Chengyong He , Wei Wang , Zhenghong Zuo
The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor that regulates various biological processes, including xenobiotic metabolism, immune response, and reproduction. Although previous studies have shown that AHR plays a role in ovarian follicle development, the precise role of oocyte-expressed AHR in female reproduction remains unclear. In this study, oocyte-specific Ahr knockout (cKO) mice generated by crossing the Ahrflox/flox (Ahrfl/fl) and Gdf9-cre transgenic mouse strains were used to answer this open question. The cKO female mice exhibited a disrupted estrous cyclicity and subfertility. Histological analyses demonstrated that oocyte AHR loss reduces the number of primary follicles while increasing the number of secondary follicles and corpus lutea in mouse ovary. Hormonal analysis revealed decreased serum estradiol and follicle-stimulating hormone, indicating a disruption of the hypothalamic-pituitary-gonadal axis in cKO mice. TUNEL and Western blotting results demonstrate that deletion of oocyte AHR also results in increased apoptosis in ovarian granulosa cells (GCs), downregulated expression of Gdf9 and Bmp15 in oocytes, and disrupted bidirectional oocyte-GC communication. In conclusion, our findings reveal that the aryl hydrocarbon receptor plays a role beyond sensing environmental chemicals and endogenous compounds and underscore a critical role of oocyte-expressed Ahr in maintain follicle development, ovarian function, and female reproductive health.
{"title":"Oocyte-specific Ahr deletion disrupts folliculogenesis and female fertility in mice","authors":"Pengtao Chen , Jinpeng Ruan , Fanzheng Xue , Xuejuan Dai , Chen Tang , Mingyue Chen , Nengming Xiao , Zhijian Cai , Chunyan Yang , Chengyong He , Wei Wang , Zhenghong Zuo","doi":"10.1016/j.mce.2025.112656","DOIUrl":"10.1016/j.mce.2025.112656","url":null,"abstract":"<div><div>The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor that regulates various biological processes, including xenobiotic metabolism, immune response, and reproduction. Although previous studies have shown that AHR plays a role in ovarian follicle development, the precise role of oocyte-expressed AHR in female reproduction remains unclear. In this study, oocyte-specific <em>Ahr</em> knockout (cKO) mice generated by crossing the <em>Ahr</em> <sup><em>flox/flox</em></sup> (<em>Ahr</em> <sup><em>fl/fl</em></sup>) and <em>Gdf9-cre</em> transgenic mouse strains were used to answer this open question. The cKO female mice exhibited a disrupted estrous cyclicity and subfertility. Histological analyses demonstrated that oocyte AHR loss reduces the number of primary follicles while increasing the number of secondary follicles and corpus lutea in mouse ovary. Hormonal analysis revealed decreased serum estradiol and follicle-stimulating hormone, indicating a disruption of the hypothalamic-pituitary-gonadal axis in cKO mice. TUNEL and Western blotting results demonstrate that deletion of oocyte AHR also results in increased apoptosis in ovarian granulosa cells (GCs), downregulated expression of <em>Gdf9</em> and <em>Bmp15</em> in oocytes<em>,</em> and disrupted bidirectional oocyte-GC communication. In conclusion, our findings reveal that the aryl hydrocarbon receptor plays a role beyond sensing environmental chemicals and endogenous compounds and underscore a critical role of oocyte-expressed Ahr in maintain follicle development, ovarian function, and female reproductive health.</div></div>","PeriodicalId":18707,"journal":{"name":"Molecular and Cellular Endocrinology","volume":"609 ","pages":"Article 112656"},"PeriodicalIF":3.6,"publicationDate":"2025-09-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145058593","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-09-04DOI: 10.1016/j.mce.2025.112655
John Dou , Soundara Viveka Thangaraj , Yiran Zhou , Vasantha Padmanabhan , Kelly Bakulski
Steroid hormones are integral to pregnancy and fetal development, regulating processes such as metabolism, inflammation, and immune responses. Excessive prenatal steroid exposure, through lifestyle choices or environmental chemicals, can lead to metabolic dysfunctions in offspring. The research focuses on how exposure to testosterone (T) and bisphenol A (BPA) affects the liver's DNA methylome, a key component of the epigenome influencing long-term health. Using Suffolk sheep, the study involved two cohorts: one exposed to prenatal-T and the other to prenatal-BPA. Whole genome bisulfite sequencing was employed to map DNA methylation across over 22 million CpG sites. Regions with p-value<10−4 and a magnitude of difference of at least 5 % methylation between groups were considered differentially methylated. Results demonstrated substantial differential methylation in the liver tissues due to both treatments, with prenatal-T causing unique epigenetic modifications distinct from those induced by prenatal-BPA. Specifically, prenatal-T treatment resulted in 53 differentially methylated regions (DMRs), of which 31 were located in gene regions, including exons. Prenatal-BPA exposure led to 32 DMRs, with 22 associated with gene regions. These modifications were associated with genes governing lipid and glucose metabolism, potentially underlying the observed metabolic disruptions such as insulin resistance and dyslipidemia. Pathway analysis revealed that genes differentially methylated due to prenatal-T were involved in cellular organization, while those affected by prenatal-BPA were enriched in signal regulation pathways. The findings underscore how prenatal exposure to steroid excess and steroid-mimics influence epigenetic landscapes, contributing to metabolic disease programming.
{"title":"Developmental programming: Differing impact of prenatal testosterone and prenatal bisphenol-A -treatment on hepatic methylome in female sheep","authors":"John Dou , Soundara Viveka Thangaraj , Yiran Zhou , Vasantha Padmanabhan , Kelly Bakulski","doi":"10.1016/j.mce.2025.112655","DOIUrl":"10.1016/j.mce.2025.112655","url":null,"abstract":"<div><div>Steroid hormones are integral to pregnancy and fetal development, regulating processes such as metabolism, inflammation, and immune responses. Excessive prenatal steroid exposure, through lifestyle choices or environmental chemicals, can lead to metabolic dysfunctions in offspring. The research focuses on how exposure to testosterone (T) and bisphenol A (BPA) affects the liver's DNA methylome, a key component of the epigenome influencing long-term health. Using Suffolk sheep, the study involved two cohorts: one exposed to prenatal-T and the other to prenatal-BPA. Whole genome bisulfite sequencing was employed to map DNA methylation across over 22 million CpG sites. Regions with p-value<10<sup>−4</sup> and a magnitude of difference of at least 5 % methylation between groups were considered differentially methylated. Results demonstrated substantial differential methylation in the liver tissues due to both treatments, with prenatal-T causing unique epigenetic modifications distinct from those induced by prenatal-BPA. Specifically, prenatal-T treatment resulted in 53 differentially methylated regions (DMRs), of which 31 were located in gene regions, including exons. Prenatal-BPA exposure led to 32 DMRs, with 22 associated with gene regions. These modifications were associated with genes governing lipid and glucose metabolism, potentially underlying the observed metabolic disruptions such as insulin resistance and dyslipidemia. Pathway analysis revealed that genes differentially methylated due to prenatal-T were involved in cellular organization, while those affected by prenatal-BPA were enriched in signal regulation pathways. The findings underscore how prenatal exposure to steroid excess and steroid-mimics influence epigenetic landscapes, contributing to metabolic disease programming.</div></div>","PeriodicalId":18707,"journal":{"name":"Molecular and Cellular Endocrinology","volume":"609 ","pages":"Article 112655"},"PeriodicalIF":3.6,"publicationDate":"2025-09-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145008373","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-09-01DOI: 10.1016/j.mce.2025.112644
Lingjuan Wang, Sijin OuYang, Bin Li, Na Kong
RNA-binding proteins (RBPs) are critical regulators of post-transcriptional gene expression and RNA processing during mammalian oocyte development. SERPINE1 mRNA-binding protein 1 (SERBP1), a conserved RNA-binding protein (RBP), exhibits prominent expression in the female reproductive system and throughout oogenesis. Conditional deletion of Serbp1 using oocyte-specific Zp3/Gdf9-Cre drivers resulted in arrested oocyte growth, female infertility, and failure of blastocyst formation from two-cell embryos. Phenotypic analysis revealed spindle assembly defects, impaired asymmetric division, and compromised meiotic competence in oocytes. Notably, Serbp1 ablation also induced granulosa cell apoptosis and elevated Erk1/2 phosphorylation levels, indicating dysregulation of somatic microenvironment. Furthermore, conditional knockout mice exhibited prolonged diestrus cycles. Collectively, these findings demonstrate that SERBP1 coordinates essential RNA-regulatory functions for oocyte developmental competence through both cell-autonomous mechanisms and somatic-germline crosstalk.
{"title":"RNA binding protein SERBP1 is indispensable for oocyte development and maturation in mice.","authors":"Lingjuan Wang, Sijin OuYang, Bin Li, Na Kong","doi":"10.1016/j.mce.2025.112644","DOIUrl":"https://doi.org/10.1016/j.mce.2025.112644","url":null,"abstract":"<p><p>RNA-binding proteins (RBPs) are critical regulators of post-transcriptional gene expression and RNA processing during mammalian oocyte development. SERPINE1 mRNA-binding protein 1 (SERBP1), a conserved RNA-binding protein (RBP), exhibits prominent expression in the female reproductive system and throughout oogenesis. Conditional deletion of Serbp1 using oocyte-specific Zp3/Gdf9-Cre drivers resulted in arrested oocyte growth, female infertility, and failure of blastocyst formation from two-cell embryos. Phenotypic analysis revealed spindle assembly defects, impaired asymmetric division, and compromised meiotic competence in oocytes. Notably, Serbp1 ablation also induced granulosa cell apoptosis and elevated Erk1/2 phosphorylation levels, indicating dysregulation of somatic microenvironment. Furthermore, conditional knockout mice exhibited prolonged diestrus cycles. Collectively, these findings demonstrate that SERBP1 coordinates essential RNA-regulatory functions for oocyte developmental competence through both cell-autonomous mechanisms and somatic-germline crosstalk.</p>","PeriodicalId":18707,"journal":{"name":"Molecular and Cellular Endocrinology","volume":" ","pages":"112644"},"PeriodicalIF":3.6,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144992903","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}