Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (696)

Search Parameters:
Keywords = estrogen receptor signaling

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
18 pages, 1241 KiB  
Review
PCOS and the Genome: Is the Genetic Puzzle Still Worth Solving?
by Mario Palumbo, Luigi Della Corte, Dario Colacurci, Mario Ascione, Giuseppe D’Angelo, Giorgio Maria Baldini, Pierluigi Giampaolino and Giuseppe Bifulco
Biomedicines 2025, 13(8), 1912; https://doi.org/10.3390/biomedicines13081912 - 5 Aug 2025
Abstract
Background: Polycystic ovary syndrome (PCOS) is a complex and multifactorial disorder affecting reproductive, endocrine, and metabolic functions in women of reproductive age. While environmental and lifestyle factors play a role, increasing evidence highlights the contribution of genetic and epigenetic mechanisms to its pathogenesis. [...] Read more.
Background: Polycystic ovary syndrome (PCOS) is a complex and multifactorial disorder affecting reproductive, endocrine, and metabolic functions in women of reproductive age. While environmental and lifestyle factors play a role, increasing evidence highlights the contribution of genetic and epigenetic mechanisms to its pathogenesis. Objective: This narrative review aims to provide an updated overview of the current evidence regarding the role of genetic variants, gene expression patterns, and epigenetic modifications in the etiopathogenesis of PCOS, with a focus on their impact on ovarian function, fertility, and systemic alterations. Methods: A comprehensive search was conducted across MEDLINE, EMBASE, PubMed, Web of Science, and the Cochrane Library using MeSH terms including “PCOS”, “Genes involved in PCOS”, and “Etiopathogenesis of PCOS” from January 2015 to June 2025. The selection process followed the SANRA quality criteria for narrative reviews. Seventeen studies published in English were included, focusing on original data regarding gene expression, polymorphisms, and epigenetic changes associated with PCOS. Results: The studies analyzed revealed a wide array of molecular alterations in PCOS, including the dysregulation of SIRT and estrogen receptor genes, altered transcriptome profiles in cumulus cells, and the involvement of long non-coding RNAs and circular RNAs in granulosa cell function and endometrial receptivity. Epigenetic mechanisms such as the DNA methylation of TGF-β1 and inflammation-related signaling pathways (e.g., TLR4/NF-κB/NLRP3) were also implicated. Some genetic variants—particularly in DENND1A, THADA, and MTNR1B—exhibit signs of positive evolutionary selection, suggesting possible ancestral adaptive roles. Conclusions: PCOS is increasingly recognized as a syndrome with a strong genetic and epigenetic background. The identification of specific molecular signatures holds promise for the development of personalized diagnostic markers and therapeutic targets. Future research should focus on large-scale genomic studies and functional validation to better understand gene–environment interactions and their influence on phenotypic variability in PCOS. Full article
Show Figures

Figure 1

30 pages, 4011 KiB  
Article
Multitarget Design of Steroidal Inhibitors Against Hormone-Dependent Breast Cancer: An Integrated In Silico Approach
by Juan Rodríguez-Macías, Oscar Saurith-Coronell, Carlos Vargas-Echeverria, Daniel Insuasty Delgado, Edgar A. Márquez Brazón, Ricardo Gutiérrez De Aguas, José R. Mora, José L. Paz and Yovanni Marrero-Ponce
Int. J. Mol. Sci. 2025, 26(15), 7477; https://doi.org/10.3390/ijms26157477 (registering DOI) - 2 Aug 2025
Viewed by 254
Abstract
Hormone-dependent breast cancer, particularly in its treatment-resistant forms, remains a significant therapeutic challenge. In this study, we applied a fully computational strategy to design steroid-based compounds capable of simultaneously targeting three key receptors involved in disease progression: progesterone receptor (PR), estrogen receptor alpha [...] Read more.
Hormone-dependent breast cancer, particularly in its treatment-resistant forms, remains a significant therapeutic challenge. In this study, we applied a fully computational strategy to design steroid-based compounds capable of simultaneously targeting three key receptors involved in disease progression: progesterone receptor (PR), estrogen receptor alpha (ER-α), and HER2. Using a robust 3D-QSAR model (R2 = 0.86; Q2_LOO = 0.86) built from 52 steroidal structures, we identified molecular features associated with high anticancer potential, specifically increased polarizability and reduced electronegativity. From a virtual library of 271 DFT-optimized analogs, 31 compounds were selected based on predicted potency (pIC50 > 7.0) and screened via molecular docking against PR (PDB 2W8Y), HER2 (PDB 7JXH), and ER-α (PDB 6VJD). Seven candidates showed strong binding affinities (ΔG ≤ −9 kcal/mol for at least two targets), with Estero-255 emerging as the most promising. This compound demonstrated excellent conformational stability, a robust hydrogen-bonding network, and consistent multitarget engagement. Molecular dynamics simulations over 100 nanoseconds confirmed the structural integrity of the top ligands, with low RMSD values, compact radii of gyration, and stable binding energy profiles. Key interactions included hydrophobic contacts, π–π stacking, halogen–π interactions, and classical hydrogen bonds with conserved residues across all three targets. These findings highlight Estero-255, alongside Estero-261 and Estero-264, as strong multitarget candidates for further development. By potentially disrupting the PI3K/AKT/mTOR signaling pathway, these compounds offer a promising strategy for overcoming resistance in hormone-driven breast cancer. Experimental validation, including cytotoxicity assays and ADME/Tox profiling, is recommended to confirm their therapeutic potential. Full article
Show Figures

Graphical abstract

16 pages, 2106 KiB  
Article
ERα36 Promotes MDR1-Mediated Adriamycin Resistance via Non-Genomic Signaling in Triple-Negative Breast Cancer
by Muslimbek Mukhammad Ugli Poyonov, Anh Thi Ngoc Bui, Seung-Yeon Lee, Gi-Ho Lee and Hye-Gwang Jeong
Int. J. Mol. Sci. 2025, 26(15), 7200; https://doi.org/10.3390/ijms26157200 - 25 Jul 2025
Viewed by 188
Abstract
Drug resistance remains a critical barrier to effective treatment in several cancers, particularly triple-negative breast cancer (TNBC). Estrogen receptor α36 (ERα36), a variant of the estrogen receptor in ER-negative breast cancer cells, plays important roles in cancer cell proliferation. We investigated the role [...] Read more.
Drug resistance remains a critical barrier to effective treatment in several cancers, particularly triple-negative breast cancer (TNBC). Estrogen receptor α36 (ERα36), a variant of the estrogen receptor in ER-negative breast cancer cells, plays important roles in cancer cell proliferation. We investigated the role of ERα36 in regulating multidrug resistance protein 1 (MDR1) in MDA-MB-231 human breast cancer cells. The activation of ERα36 by BSA-conjugated estradiol (BSA-E2) increased cell viability under Adriamycin exposure, suggesting its involvement in promoting drug resistance. BSA-E2 treatment significantly reduced the intracellular rhodamine-123 levels by activating the MDR1 efflux function, which was linked to increased MDR1 transcription and protein expression. The mechanical ERα36-mediated BSA-E2-induced activation of EGFR and downstream signaling via c-Src led to an activation of the Akt/ERK pathways and transcription factors, NF-κB and CREB. Additionally, ERα36 is involved in activating Wnt/β-catenin pathways to induce MDR1 expression. The silencing of ERα36 inhibited the BSA-E2-induced phosphorylation of Akt and ERK, thereby reducing MDR1 expression via downregulation of NF-κB and CREB as well as Wnt/β-catenin signaling. These findings demonstrated that ERα36 promotes MDR1 expression through multiple non-genomic signaling cascades, including Akt/ERK-NF-κB/CREB and Wnt/β-catenin pathways, and highlight the role of ERα36 as a promising target to enhance chemotherapeutic efficacy in TNBC. Full article
(This article belongs to the Special Issue Drug Resistance Mechanisms in Human Cancer Cells to Anticancer Drugs)
Show Figures

Figure 1

31 pages, 4221 KiB  
Article
Estradiol Downregulates MicroRNA-193a to Mediate Its Angiogenic Actions
by Lisa Rigassi, Mirel Adrian Popa, Ruth Stiller, Brigitte Leeners, Marinella Rosselli and Raghvendra Krishna Dubey
Cells 2025, 14(15), 1134; https://doi.org/10.3390/cells14151134 - 23 Jul 2025
Cited by 1 | Viewed by 348
Abstract
Estrogens regulate many physiological processes in the human body, including the cardiovascular system. Importantly, Estradiol (E2) exerts its vascular protective actions, in part, by promoting endothelial repair via induction of endothelial cell (EC) proliferation, migration and angiogenesis. Recent evidence that microRNAs (miRNAs) play [...] Read more.
Estrogens regulate many physiological processes in the human body, including the cardiovascular system. Importantly, Estradiol (E2) exerts its vascular protective actions, in part, by promoting endothelial repair via induction of endothelial cell (EC) proliferation, migration and angiogenesis. Recent evidence that microRNAs (miRNAs) play an important role in vascular health and disease as well as in regulating Estrogen actions in many cell types. We hypothesize that E2 may mediate its vascular protective actions via the regulation of miRNAs. Following initial screening, we found that E2 downregulates the levels of miR-193a-3p in ECs. Moreover, miR-193a-3p downregulation by miR-193a-3p-antimir mimicked the effects as E2 on EC growth, migration, and capillary formation. Restoring miR-193a-3p levels with mimics after E2 treatment abrogated the vasculogenic actions of E2, suggesting a key role of miR-193a-3p in E2-mediated EC-growth-promoting effects. We further investigated the cellular mechanisms involved and found that miR-193a-3p inhibits angiogenesis by blocking phosphoinositide-3-kinase (PI3K)/Akt-vascular endothelial growth factor (VEGF) and Activin receptor-like kinase 1 (ALK1)/SMAD1/5/8 signaling in ECs, both pathways that are important in E2-mediated vascular protection. Additionally, using reverse transcription polymerase chain reaction (RT-PCR), we demonstrate that E2 downregulates miR-193a-3p in ECs via Estrogen Receptor (ER)α, but not ERβ or G protein-coupled estrogen receptor (GPER). Moreover, these actions occur post-transcriptionally, as the expression of pri-miR-193a-3p was not affected. The anti-angiogenic actions of miR-193a-3p were also observed in in vivo Matrigel implant-based capillary formation studies in ovariectomized mice where E2 induced capillary formation, and these effects were abrogated in the presence of miR-193a-3p, but not in the control mimic. Assessment of miR-193a-3p levels in plasma collected from in vitro fertilization (IVF) subjects with low and high E2 levels showed significantly lower miR-193a-3p levels in responders during the high E2 period. Hence, our findings provide the first evidence that miR-193a-3p mimic inhibits angiogenesis whereas its antimir is angiogenic. Importantly, E2 mediates its regenerative actions on ECs/capillary formation by downregulating endogenous miR-193a-3p expression. Both miR-193a-3p mimic or antimir may represent important therapeutic molecules to prevent or to induce endothelial function in treating pathophysiologies associated with capillary growth. Full article
Show Figures

Graphical abstract

32 pages, 10235 KiB  
Article
Estradiol Downregulates MicroRNA-193a to Mediate Its Anti-Mitogenic Actions on Human Coronary Artery Smooth Muscle Cell Growth
by Lisa Rigassi, Marinella Rosselli, Brigitte Leeners, Mirel Adrian Popa and Raghvendra Krishna Dubey
Cells 2025, 14(15), 1132; https://doi.org/10.3390/cells14151132 - 23 Jul 2025
Viewed by 307
Abstract
The abnormal growth of smooth muscle cells (SMCs) contributes to the vascular remodeling associated with coronary artery disease, a leading cause of death in women. Estradiol (E2) mediates cardiovascular protective actions, in part, by inhibiting the abnormal growth (proliferation and migration) of SMCs [...] Read more.
The abnormal growth of smooth muscle cells (SMCs) contributes to the vascular remodeling associated with coronary artery disease, a leading cause of death in women. Estradiol (E2) mediates cardiovascular protective actions, in part, by inhibiting the abnormal growth (proliferation and migration) of SMCs through various mechanism. Since microRNAs (miRNAs) play a major role in regulating cell growth and vascular remodeling, we hypothesize that miRNAs may mediate the protective actions of E2. Following preliminary leads from E2-regulated miRNAs, we found that platelet-derived growth factor (PDGF)-BB-induced miR-193a in SMCs is downregulated by E2 via estrogen receptor (ER)α, but not the ERβ or G-protein-coupled estrogen receptor (GPER). Importantly, miR-193a is actively involved in regulating SMC functions. The ectopic expression of miR-193a induced vascular SMC proliferation and migration, while its suppression with antimir abrogated PDGF-BB-induced growth, effects that were similar to E2. Importantly, the restoration of miR-193a abrogated the anti-mitogenic actions of E2 on PDGF-BB-induced growth, suggesting a key role of miR-193a in mediating the growth inhibitory actions of E2 in vascular SMCs. E2-abrogated PDGF-BB, but not miR-193a, induced SMC growth, suggesting that E2 blocks the PDGF-BB-induced miR-193a formation to mediate its anti-mitogenic actions. Interestingly, the PDGF-BB-induced miR-193a formation in SMCs was also abrogated by 2-methoxyestradiol (2ME), an endogenous E2 metabolite that inhibits SMC growth via an ER-independent mechanism. Furthermore, we found that miR-193a induces SMC growth by activating the phosphatidylinositol 3-kinases (PI3K)/Akt signaling pathway and promoting the G1 to S phase progression of the cell cycle, by inducing Cyclin D1, Cyclin Dependent Kinase 4 (CDK4), Cyclin E, and proliferating-cell-nuclear-antigen (PCNA) expression and Retinoblastoma-protein (RB) phosphorylation. Importantly, in mice, treatment with miR-193a antimir, but not its control, prevented cuff-induced vascular remodeling and significantly reducing the vessel-wall-to-lumen ratio in animal models. Taken together, our findings provide the first evidence that miR-193a promotes SMC proliferation and migration and may play a key role in PDGF-BB-induced vascular remodeling/occlusion. Importantly, E2 prevents PDGF-BB-induced SMC growth by downregulating miR-193a formation in SMCs. Since, miR-193a antimir prevents SMC growth as well as cuff-induced vascular remodeling, it may represent a promising therapeutic molecule against cardiovascular disease. Full article
Show Figures

Graphical abstract

14 pages, 814 KiB  
Review
Menopause as a Critical Turning Point in Lipedema: The Estrogen Receptor Imbalance, Intracrine Estrogen, and Adipose Tissue Dysfunction Model
by Diogo Pinto da Costa Viana, Lucas Caseri Câmara and Robinson Borges Palau
Int. J. Mol. Sci. 2025, 26(15), 7074; https://doi.org/10.3390/ijms26157074 - 23 Jul 2025
Viewed by 2006
Abstract
Lipedema is a chronic, estrogen-sensitive adipose tissue disorder characterized by disproportionate subcutaneous fat accumulation, fibrosis, inflammation, and resistance to fat mobilization. Despite its high prevalence, lipedema remains poorly understood and frequently misdiagnosed. This narrative review proposes a novel pathophysiological model in which menopause [...] Read more.
Lipedema is a chronic, estrogen-sensitive adipose tissue disorder characterized by disproportionate subcutaneous fat accumulation, fibrosis, inflammation, and resistance to fat mobilization. Despite its high prevalence, lipedema remains poorly understood and frequently misdiagnosed. This narrative review proposes a novel pathophysiological model in which menopause acts as a critical turning point in the progression of lipedema, driven by estrogen receptor imbalance (ERβ predominance over ERα), intracrine estrogen excess, and adipose tissue dysfunction. We demonstrate how menopause amplifies adipose tissue dysfunction by suppressing ERα signaling; enhancing ERβ activity; and disrupting mitochondrial function, insulin sensitivity, and lipid oxidation. Concurrently, the upregulation of aromatase and 17β-HSD1, combined with the suppression of 17β-HSD2, sustains localized estradiol excess, perpetuating inflammation, fibrosis, and immune dysregulation. The molecular signature observed in lipedema closely mirrors that of other estrogen-driven gynecological disorders, such as endometriosis, adenomyosis, and uterine fibroids. Understanding these molecular mechanisms highlights the pivotal role of menopause as a catalyst for disease progression and provides a rationale for targeted therapeutic strategies, including hormonal modulation and metabolic interventions. This review reframes lipedema as an estrogen receptor-driven gynecological disorder, offering a new perspective to improve clinical recognition, diagnosis, and management of this neglected condition. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

19 pages, 1835 KiB  
Article
Transcriptome Analysis Reveals Hyperglycemic Hormone and Excitatory Amino Acid Transporter 3 Are Involved in the Thermal Adaptation of Eriocheir sinensis
by Xi Li, Runlin Zhou, Ruiqi Zhang, Zhen He, Mingzhi Zhang, Ran Li, Tong Hao and Jinsheng Sun
Fishes 2025, 10(7), 361; https://doi.org/10.3390/fishes10070361 - 21 Jul 2025
Viewed by 295
Abstract
Temperature is one of the critical factors influencing the survival, growth, and reproduction of organisms. The molting and developmental mechanisms of crustaceans are highly sensitive to temperature, yet the regulatory mechanisms underlying their thermal adaptation remain unclear. In this work, transcriptome sequencing was [...] Read more.
Temperature is one of the critical factors influencing the survival, growth, and reproduction of organisms. The molting and developmental mechanisms of crustaceans are highly sensitive to temperature, yet the regulatory mechanisms underlying their thermal adaptation remain unclear. In this work, transcriptome sequencing was performed to analyze the gene expression profiles of Eriocheir sinensis under normal temperature (22 °C) and high-temperature (27 °C and 32 °C) conditions. A total of 377 differentially expressed genes (DEGs) were identified, including 149 up-regulated and 227 down-regulated genes. Through Gene Ontology (GO) enrichment analysis of these DEGs, 11 significantly temperature-regulated signaling pathways were identified, including the estrogen and androgen receptor signaling pathways, and two neurotransmission signaling pathways. These findings suggest that temperature may influence sex regulation in E. sinensis, while the dopamine receptor and neuropeptide signaling pathways may play a role in its thermal adaptation. Further validation via RT-qPCR of DEGs involved in neurotransmission signaling pathways revealed that crustacean hyperglycemic hormone (CHH) and excitatory amino acid transporter 3 (EAA3) genes are likely involved in the thermal adaptation of E. sinensis. In addition, the hemolymph glucose levels associated with the elevated temperatures were detected and consistent variations between glucose levels and CHH expressions were found. This indicates that the eyestalk CHH is strongly correlated with the hemolymph glucose levels and likely mediates the response to temperature changes by regulating blood glucose in E. sinensis. The results of this study not only provide key molecular targets for elucidating the mechanisms by which temperature affects molting and development in E. sinensis, but also establish a theoretical foundation for further research into thermal adaptation strategies in crustaceans. Full article
(This article belongs to the Section Aquatic Invertebrates)
Show Figures

Figure 1

11 pages, 1606 KiB  
Article
Exploring the Therapeutic Potential of Estrogen-Related Receptor γ Inverse Agonists in Atopic Dermatitis-like Lesions
by Ju Hyeon Bae, Sijoon Lee, Jae-Eon Lee, Sang Kyoon Kim, Jae-Han Jeon and Yong Hyun Jeon
Int. J. Mol. Sci. 2025, 26(14), 6959; https://doi.org/10.3390/ijms26146959 - 20 Jul 2025
Viewed by 265
Abstract
Estrogen-related receptor γ (ERRγ) has been reported to regulate various inflammation-related diseases. Herein, we attempted to evaluate the effects of DN200434 as a modulator for ERRγ in mice with atopic dermatitis (AD). Levels of mRNA and protein expression for ERRγ were evaluated in [...] Read more.
Estrogen-related receptor γ (ERRγ) has been reported to regulate various inflammation-related diseases. Herein, we attempted to evaluate the effects of DN200434 as a modulator for ERRγ in mice with atopic dermatitis (AD). Levels of mRNA and protein expression for ERRγ were evaluated in normal and DNCB-induced AD-diagnosed skin. The effects of DN200434 on the chemokines, inflammatory cytokines, and AKT/MAPK/NFκB pathway signaling were investigated in TNF-α/IFN-γ-treated HaCaT cells. DNCB-induced AD mice received DN200434 intraperitoneally for 10 days. Epidermal thickness at the dorsal aspect of the inflamed skin, spleen index, serum IgE levels, and proinflammatory cytokine levels in the skin lesions were measured. Histopathological evaluations, including assessments of epidermal hyperplasia, dermal inflammation, hyperkeratosis, folliculitis, and mast cell counts, were performed to confirm diagnostic features. Significant elevations in ERRγ expression at the RNA and protein levels were observed in DNCB-induced AD lesions. DN200434 suppressed chemokine and inflammatory cytokine expression and inhibited the elevated phosphorylation levels of AKT, ERK, p38, and NFκB in TNF-α/IFN-γ-treated HaCaT cells. Treatment with DN200434 alleviated DNCB-induced AD symptoms. The histopathological score and levels of infiltrated mast cells were also markedly lower in DN200434-treated AD mice than in vehicle-treated AD mice. Consistently, DN200434 reduced the serum IgE level and mRNA expression of TNFα and IL-6 in AD-diagnosed lesions. Collectively, our findings indicated the feasibility of ERRγ as a therapeutic target for the regulation of AD and that DN200434 can be a useful therapeutic agent in treating AD. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

21 pages, 3526 KiB  
Article
Prenatal Bisphenol A Exposure Impairs Fetal Heart Development: Molecular and Structural Alterations with Sex-Specific Differences
by Alessandro Marrone, Anna De Bartolo, Vittoria Rago, Francesco Conforti, Lidia Urlandini, Tommaso Angelone, Rosa Mazza, Maurizio Mandalà and Carmine Rocca
Antioxidants 2025, 14(7), 863; https://doi.org/10.3390/antiox14070863 - 14 Jul 2025
Viewed by 439
Abstract
Cardiovascular diseases (CVDs) remain the leading cause of morbidity and mortality worldwide, with increasing evidence suggesting that their origins may lie in prenatal life. Endocrine-disrupting chemicals (EDCs), such as bisphenol A (BPA), have been implicated in the alteration of fetal programming mechanisms that [...] Read more.
Cardiovascular diseases (CVDs) remain the leading cause of morbidity and mortality worldwide, with increasing evidence suggesting that their origins may lie in prenatal life. Endocrine-disrupting chemicals (EDCs), such as bisphenol A (BPA), have been implicated in the alteration of fetal programming mechanisms that cause a predisposition to long-term cardiovascular vulnerability. However, the impact of prenatal endocrine disruption on fetal heart development and its sex-specific nature remains incompletely understood. This study investigates the molecular and structural effects of low-dose prenatal BPA exposure on fetal rat hearts. Our results reveal that BPA disrupts estrogen receptor (ER) signaling in a sex-dependent manner, with distinct alterations in ERα, ERβ, and GPER expression. BPA exposure also triggers significant inflammation, oxidative stress, and ferroptosis; this is evidenced by elevated NF-κB, IL-1β, TNF-α, and NLRP3 inflammasome activation, as well as impaired antioxidant defenses (SOD1, SOD2, CAT, and SELENOT), increased lipid peroxidation (MDA) and protein oxidation, decreased GPX4, and increased ACSL4 levels. These alterations are accompanied by increased markers of cardiac distension (ANP, BNP), extracellular matrix remodeling mediators, and pro-fibrotic regulators (Col1A1, Col3A1, TGF-β, and CTGF), with a more pronounced response in males. Histological analyses corroborated these molecular findings, revealing structural alterations as well as glycogen depletion in male fetal hearts, consistent with altered cardiac morphogenesis and metabolic stress. These effects were milder in females, reinforcing the notion of sex-specific vulnerability. Moreover, prenatal BPA exposure affected myocardial fiber architecture and vascular remodeling in a sex-dependent manner, as evidenced by reduced expression of desmin alongside increased levels of CD34 and Ki67. Overall, our findings provide novel insights into the crucial role of prenatal endocrine disruption during fetal heart development and its contribution to the early origins of CVD, underscoring the urgent need for targeted preventive strategies and further research into the functional impact of BPA-induced alterations on postnatal cardiac function and long-term disease susceptibility. Full article
Show Figures

Graphical abstract

21 pages, 3110 KiB  
Article
Long-Chain Fatty Acids Alter Estrogen Receptor Expression in Breast Cancer Cells
by Ruiko Ogata, Yi Luo, Rina Fujiwara-Tani, Rika Sasaki, Ayaka Ikemoto, Kaho Maehana, Ayaka Sasaki, Takamitsu Sasaki, Kiyomu Fujii, Hitoshi Ohmori and Hiroki Kuniyasu
Int. J. Mol. Sci. 2025, 26(14), 6722; https://doi.org/10.3390/ijms26146722 - 13 Jul 2025
Viewed by 490
Abstract
Long-chain fatty acids (LCFAs) have emerged as important regulators of cancer metabolism, but their impact on hormone receptor expression in breast cancer (BCA) remains poorly understood. In this study, we investigated the effects of five LCFAs—linoleic acid (LA), oleic acid (OA), elaidic acid [...] Read more.
Long-chain fatty acids (LCFAs) have emerged as important regulators of cancer metabolism, but their impact on hormone receptor expression in breast cancer (BCA) remains poorly understood. In this study, we investigated the effects of five LCFAs—linoleic acid (LA), oleic acid (OA), elaidic acid (EA), palmitic acid (PA), and α-linolenic acid (LNA)—on two BCA cell lines: luminal-type MCF7 and triple-negative MDA-MB-231 (MB231). All LCFAs suppressed cell viability and mitochondrial function in a dose-dependent manner, accompanied by decreased membrane potential, increased reactive oxygen species production, and a metabolic shift. Notably, OA reduced both mRNA and nuclear protein levels of estrogen receptor alpha (ERα) in MCF7 cells, leading to impaired responses to estradiol and tamoxifen. In contrast, PA induced nuclear ERα expression in MB231 cells, although ER signaling remained inactive. MicroRNA profiling revealed that OA upregulated ER-suppressive miR-22 and miR-221 in MCF7, while PA increased miR-34a in MB231, contributing to ERα induction. These findings suggest that specific LCFAs modulate ER expression through epigenetic and post-transcriptional mechanisms, altering hormonal responsiveness in BCA. Our results offer new insights into how dietary lipids may influence therapeutic efficacy and tumor behavior by regulating nuclear receptor signaling. Full article
(This article belongs to the Special Issue Molecular Mechanisms and Targeted Therapies of Breast Cancer)
Show Figures

Figure 1

19 pages, 2530 KiB  
Article
Comparative Analyses of IGF-Induced Liver Transcriptomes Reveal Genes and Signaling Pathways Associated with Ovarian Growth and Development in Golden Pompano (Trachinotus ovatus)
by Yan Wang, Charles Brighton Ndandala, Muhammad Fachri, Vicent Michael Shija, Pengfei Li and Huapu Chen
Fishes 2025, 10(7), 315; https://doi.org/10.3390/fishes10070315 - 2 Jul 2025
Viewed by 228
Abstract
Recently, China has become a hotspot for farming golden pompano (Trachinotus ovatus), a commercially valuable marine fish. The genetic mechanisms underlying ovarian development, particularly those regulated by insulin-like growth factors (IGFs), remain poorly understood. Existing research on T. ovatus has focused [...] Read more.
Recently, China has become a hotspot for farming golden pompano (Trachinotus ovatus), a commercially valuable marine fish. The genetic mechanisms underlying ovarian development, particularly those regulated by insulin-like growth factors (IGFs), remain poorly understood. Existing research on T. ovatus has focused primarily on growth metrics, developmental stages, and immune responses, leaving a critical gap in knowledge regarding the hepatic regulatory pathways activated by IGFs. In this study, differentially expressed genes (DEGs) were detected through RNA sequencing (RNA-Seq) and associated pathways in response to IGF treatment. Comparisons between the IGF1, IGF2, and IGF3 treated groups and the control revealed 113 (46 upregulated, 67 downregulated), 637 (567 upregulated, 70 downregulated), and 587 DEGs (273 upregulated, 314 downregulated), respectively. KEGG enrichment analysis highlighted key pathways that may be linked to ovarian growth and development, including biotin metabolism, biosynthesis of amino acids, drug-cytochrome p450 pathways, MAPK signaling, estrogen signaling pathways, ECM receptor interaction, steroid biosynthesis, and ovarian steroidogenesis. These findings advance our understanding of hepatic metabolic regulation in golden pompano via the IGF system and provide actionable insights for optimizing aquaculture practices and selective breeding programs for this species. Full article
(This article belongs to the Section Genetics and Biotechnology)
Show Figures

Figure 1

15 pages, 263 KiB  
Review
Challenges in Differentiating Uterine Mesenchymal Tumors—Key Diagnostic Criteria
by Karolina Daniłowska, Małgorzata Satora, Krzysztof Kułak, Anna Kułak and Rafał Tarkowski
J. Clin. Med. 2025, 14(13), 4644; https://doi.org/10.3390/jcm14134644 - 1 Jul 2025
Viewed by 433
Abstract
Background: Uterine fibroids are the most common tumors in gynecology, detected in up to 80% of patients at various points in their lives. Uterine sarcomas account for 3% to 7% of all uterine cancers. The diagnosis of uterine fibroids is possible through [...] Read more.
Background: Uterine fibroids are the most common tumors in gynecology, detected in up to 80% of patients at various points in their lives. Uterine sarcomas account for 3% to 7% of all uterine cancers. The diagnosis of uterine fibroids is possible through ultrasonography (US), but this method has many limitations. More accurate examinations include magnetic resonance imaging (MRI) and positron emission tomography (PET) scans. Methods: This study evaluates MRI and PET in differentiating uterine fibroids from sarcomas. MRI uses T2-weighted and diffusion-weighted imaging (DWI), while PET assesses metabolism and estrogen receptor activity using [18F] fluorodeoxyglucose (FDG) and 16α-[18F]-fluoro-17β-estradiol (FES). Results: MRI allows for the identification of uterine fibroids when they exhibit good delineation and low intensity in T2-weighted images and DWI. Uterine sarcoma is characterized by moderate to high signal intensity on T2-weighted imaging, irregular borders, high signal intensity at high DWI values, and a decreased apparent diffusion coefficient. PET imaging with FDG and FES is a useful tool in differentiating uterine fibroids from sarcomas. Uterine sarcomas exhibit greater FDG uptake than smooth muscle fibroids, although cases of similar uptake do occur. On the other hand, FES provides information about estrogen receptors (ERs). Conclusions: Future research should focus on conducting standardized imaging studies, which would facilitate the inclusion of larger patient cohorts. This, in turn, would enable the development of specific diagnostic guidelines, ultimately leading to more accurate diagnoses and reducing the difficulty of differentiating these tumors through imaging. Full article
30 pages, 2884 KiB  
Review
Silibinin Anticancer Effects Through the Modulation of the Tumor Immune Microenvironment in Triple-Negative Breast Cancer
by Shubham D. Mishra, Patricia Mendonca, Sukhmandeep Kaur and Karam F. A. Soliman
Int. J. Mol. Sci. 2025, 26(13), 6265; https://doi.org/10.3390/ijms26136265 - 28 Jun 2025
Viewed by 1052
Abstract
Triple-negative breast cancer (TNBC), characterized by the absence of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2), remains a therapeutic challenge due to its aggressive nature, limited treatment options, and high recurrence rates. Current therapies, including chemotherapy [...] Read more.
Triple-negative breast cancer (TNBC), characterized by the absence of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2), remains a therapeutic challenge due to its aggressive nature, limited treatment options, and high recurrence rates. Current therapies, including chemotherapy and immune checkpoint inhibitors, face resistance driven by tumor heterogeneity, immunosuppressive signaling, and dysregulated redox pathways. This review explores silibinin’s potential to modulate the tumor immune microenvironment (TIME) and overcome therapeutic resistance in TNBC. Silibinin exerts multifaceted anticancer effects by suppressing PD-L1 expression through the inhibition of JAK/STAT3 signaling and MUC1-C interaction, attenuating NF-κB-driven inflammation, and downregulating CCL2-mediated recruitment of tumor-associated macrophages (TAMs). Additionally, silibinin disrupts redox adaptation by targeting the Nrf2-EGFR-MYC-TXNIP axis, enhancing oxidative stress and chemosensitivity. Preclinical studies highlight its ability to inhibit epithelial–mesenchymal transition (EMT), reduce cancer stem cell (CSC) populations, and synergize with existing therapies like PD-1 inhibitors. Despite its low bioavailability, advanced formulations such as liposomes and nanoparticles show promise in improving delivery and efficacy. By reshaping TIME through dual antioxidant and immunomodulatory mechanisms, silibinin emerges as a viable adjunct therapy to reverse immunosuppression and chemoresistance in TNBC. Full article
(This article belongs to the Special Issue Bioactive Compounds and Their Anticancer Effects)
Show Figures

Figure 1

28 pages, 7888 KiB  
Article
Estradiol Prevents Amyloid Beta-Induced Mitochondrial Dysfunction and Neurotoxicity in Alzheimer’s Disease via AMPK-Dependent Suppression of NF-κB Signaling
by Pranav Mishra, Ehsan K. Esfahani, Paul Fernyhough and Benedict C. Albensi
Int. J. Mol. Sci. 2025, 26(13), 6203; https://doi.org/10.3390/ijms26136203 - 27 Jun 2025
Viewed by 704
Abstract
Alzheimer’s disease (AD), the most common form of dementia, is a progressive neurodegenerative disorder characterized by memory loss and cognitive decline. In addition to its two major pathological hallmarks, extracellular amyloid beta (Aβ) plaques and intracellular neurofibrillary tangles (NFTs), recent evidence highlights the [...] Read more.
Alzheimer’s disease (AD), the most common form of dementia, is a progressive neurodegenerative disorder characterized by memory loss and cognitive decline. In addition to its two major pathological hallmarks, extracellular amyloid beta (Aβ) plaques and intracellular neurofibrillary tangles (NFTs), recent evidence highlights the critical roles of mitochondrial dysfunction and neuroinflammation in disease progression. Aβ impairs mitochondrial function, which, in part, can subsequently trigger inflammatory cascades, creating a vicious cycle of neuronal damage. Estrogen receptors (ERs) are widely expressed throughout the brain, and the sex hormone 17β-estradiol (E2) exerts neuroprotection through both anti-inflammatory and mitochondrial mechanisms. While E2 exhibits neuroprotective properties, its mechanisms against Aβ toxicity remain incompletely understood. In this study, we investigated the neuroprotective effects of E2 against Aβ-induced mitochondrial dysfunction and neuroinflammation in primary cortical neurons, with a particular focus on the role of AMP-activated protein kinase (AMPK). We found that E2 treatment significantly increased phosphorylated AMPK and upregulated the expression of mitochondrial biogenesis regulator peroxisome proliferator-activated receptor gamma coactivator-1 α (PGC-1α), leading to improved mitochondrial respiration. In contrast, Aβ suppressed AMPK and PGC-1α signaling, impaired mitochondrial function, activated the pro-inflammatory nuclear factor kappa-light-chain enhancer of activated B cells (NF-κB), and reduced neuronal viability. E2 pretreatment also rescued Aβ-induced mitochondrial dysfunction, suppressed NF-κB activation, and, importantly, prevented the decline in neuronal viability. However, the pharmacological inhibition of AMPK using Compound C (CC) abolished these protective effects, resulting in mitochondrial collapse, elevated inflammation, and cell death, highlighting AMPK’s critical role in mediating E2’s actions. Interestingly, while NF-κB inhibition using BAY 11-7082 partially restored mitochondrial respiration, it failed to prevent Aβ-induced cytotoxicity, suggesting that E2’s full neuroprotective effects rely on broader AMPK-dependent mechanisms beyond NF-κB suppression alone. Together, these findings establish AMPK as a key mediator of E2’s protective effects against Aβ-driven mitochondrial dysfunction and neuroinflammation, providing new insights into estrogen-based therapeutic strategies for AD. Full article
(This article belongs to the Section Molecular Neurobiology)
Show Figures

Graphical abstract

20 pages, 3210 KiB  
Article
High-Calorie Diet Exacerbates the Crosstalk Between Gestational Diabetes and Youth-Onset Diabetes in Female Offspring Through Disrupted Estrogen Signaling
by Xinyu Jia, Xiangju Cao, Yuan Wang, Shuai Yang and Lixia Ji
Nutrients 2025, 17(13), 2128; https://doi.org/10.3390/nu17132128 - 26 Jun 2025
Viewed by 383
Abstract
Background/Objectives: Recent global trends highlight a concerning rise in youth-onset type 2 diabetes (YOT2D), with a marked female preponderance. We aim to explore the crosstalk between gestational diabetes mellitus (GDM) and YOT2D in female offspring. Methods: In vivo, GDM mice were [...] Read more.
Background/Objectives: Recent global trends highlight a concerning rise in youth-onset type 2 diabetes (YOT2D), with a marked female preponderance. We aim to explore the crosstalk between gestational diabetes mellitus (GDM) and YOT2D in female offspring. Methods: In vivo, GDM mice were induced by Western diet (WD), and their female offspring were fed normal diet or WD within 3 to 8 weeks. We continuously detected the glucose metabolism disorders, serum estradiol level (ELISA), and the process of ovarian maturation. Meanwhile, the dynamic changes in ERα and insulin signal in liver were monitored (qPCR, Western blot). In vitro, LO2 cells were treated with estradiol or ER antagonist BHPI to further explore the mechanism. Results: More than 85% of pregnant mice induced by WD were GDM models. The serum estradiol level in GDM offspring mice was decreased during sexual maturation, accompanied by marked oral glucose intolerance, insulin resistance, and even diabetes. The advance of sexual maturation and the decrease in serum estradiol in GDM offspring were mainly due to the downregulation of CYP19A1 in the ovaries, the reduced area of secondary follicles, and the increased number of atresia follicles, which could be greatly worsened by WD. Furthermore, GDM suppressed the protein levels of ERα, p-IRS-1, and p-Akt in liver tissue, that is, estrogen signals and insulin signaling were simultaneously weakened. WD further exacerbated the above changes. In vitro, estradiol upregulated the protein levels of ERα, p-IRS-1, and p-Akt in LO2 cells, while BHPI inhibited these changes. Conclusions: Maternal GDM promotes a high incidence of YOT2D in female offspring by affecting ovarian maturation, and a high-calorie diet exacerbates this process. Full article
(This article belongs to the Section Nutrition in Women)
Show Figures

Figure 1

Back to TopTop