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
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
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (1,427)

Search Parameters:
Keywords = ovarian proteins

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
18 pages, 1425 KiB  
Review
TMCO1 as an Endoplasmic Reticulum Calcium Load-Activated Channel: Mechanisms and Disease Implications
by Jingbo Wang, Panpan Zhu, Zhuohang Li, Xiaohui Su, Mingzhu Qi, Aimin Zhou and Xiangying Kong
Biomolecules 2025, 15(8), 1200; https://doi.org/10.3390/biom15081200 - 20 Aug 2025
Viewed by 194
Abstract
Calcium ions (Ca2+) play a vital role in many biological processes. Transmembrane and coiled-coil domain 1 (TMCO1) has been characterized as an endoplasmic reticulum (ER) transmembrane protein in recent years. It keeps the cytoplasm and ER’s Ca2+ homeostasis stable by [...] Read more.
Calcium ions (Ca2+) play a vital role in many biological processes. Transmembrane and coiled-coil domain 1 (TMCO1) has been characterized as an endoplasmic reticulum (ER) transmembrane protein in recent years. It keeps the cytoplasm and ER’s Ca2+ homeostasis stable by acting as a novel calcium channel. Studies from different laboratories have revealed that the mutation or deficiency of TMCO1 is closely correlated with several diseases, including cerebro-facio-thoracic dysplasia (CFTD), glaucoma, premature ovarian failure (POF), osteoporosis, and cancer. Here, we review the characteristics of TMCO1 and its involvement in related diseases, which may provide useful information for developing therapeutic strategies for these diseases, as well as promote further research on this protein. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

21 pages, 2581 KiB  
Review
Post-Translational Modifications in Mammalian Folliculogenesis and Ovarian Pathologies
by Dake Chen, Yue Feng, Junjing Wu, Jiawei Zhou, Zipeng Li, Mu Qiao, Tong Chen, Zhong Xu, Xianwen Peng and Shuqi Mei
Cells 2025, 14(16), 1292; https://doi.org/10.3390/cells14161292 - 20 Aug 2025
Viewed by 172
Abstract
Post-translational modifications (PTMs) of proteins, as the core mechanism for dynamically regulating follicular development, affect the maintenance of mammalian fertility by precisely coordinating granulosa cell–oocyte interaction, metabolic reprogramming, and epigenetic remodeling. Dysregulation of these modifications directly contributes to major reproductive diseases, including polycystic [...] Read more.
Post-translational modifications (PTMs) of proteins, as the core mechanism for dynamically regulating follicular development, affect the maintenance of mammalian fertility by precisely coordinating granulosa cell–oocyte interaction, metabolic reprogramming, and epigenetic remodeling. Dysregulation of these modifications directly contributes to major reproductive diseases, including polycystic ovary syndrome (PCOS) and premature ovarian insufficiency (POI). Post-translational modifications regulate follicular development through intricate mechanisms. Thus, this review systematically synthesizes recent advances in PTMs, encompassing traditional ones such as phosphorylation, ubiquitination, and acetylation, alongside emerging modifications including lactylation, SUMOylation, and ISGylation, thereby constructing a more comprehensive PTM landscape of follicular development. Furthermore, this study dissects the molecular interaction networks of these PTMs during follicular activation, maturation, and ovulation, and uncovers the common mechanisms through which PTM dysregulation contributes to pathological conditions, including hyperandrogenism in PCOS and follicular depletion in POI. Finally, this review ultimately provides a theoretical basis for improving livestock reproductive efficiency and precise intervention in clinical ovarian diseases. Full article
Show Figures

Figure 1

25 pages, 706 KiB  
Review
The Roles of Non-Coding RNAs in the Pathogenesis of Uterine Fibroids
by Drake Boos, Tsai-Der Chuang and Omid Khorram
Cells 2025, 14(16), 1290; https://doi.org/10.3390/cells14161290 - 20 Aug 2025
Viewed by 197
Abstract
Uterine fibroids are benign smooth muscle tumors that affect ~70% of women, with Black women being affected at a disproportionate rate. The growth of these tumors is driven by estrogen and progesterone. Driver mutations in genes such as MED12, HMGA2, and FH also [...] Read more.
Uterine fibroids are benign smooth muscle tumors that affect ~70% of women, with Black women being affected at a disproportionate rate. The growth of these tumors is driven by estrogen and progesterone. Driver mutations in genes such as MED12, HMGA2, and FH also play roles in the development and growth of fibroids. Despite their high prevalence, the pathogenesis of fibroids remains largely unknown, leading to a lack of effective therapeutic options. Non-coding RNAs (ncRNAs), including miRNAs (e.g., miR-21, miR-29, miR-200), lncRNAs (e.g., H19, MIAT, XIST), and circRNAs, are important regulatory RNAs that are becoming increasingly implicated in the aberrant expression of protein-coding genes functionally associated with ECM production, cell proliferation, apoptosis, and inflammation in fibroids. Race/ethnicity, MED12 mutations, and ovarian steroids influence the expression of ncRNA expression, further implicating their relevance to fibroid pathogenesis. Therapeutic targeting of these dysregulated ncRNAs in fibroids could enable more precise and individualized non-hormonal-based treatment for this common gynecologic tumor. Full article
Show Figures

Figure 1

15 pages, 3437 KiB  
Article
Adaptation of the Protocol for the Isolation of Biotinylated Protein Complexes for Drosophila melanogaster Tissues
by Igor A. Shokodko, Rustam H. Ziganshin and Nadezhda E. Vorobyeva
Int. J. Mol. Sci. 2025, 26(16), 8009; https://doi.org/10.3390/ijms26168009 - 19 Aug 2025
Viewed by 169
Abstract
Proximity biotinylation, which utilizes various biotin ligating enzymes (BioID, TurboID, etc.), is widely used as a powerful tool for identifying novel protein–protein interactions. However, this method has a significant limitation: the use of streptavidin on beads for enriching biotinylated proteins often results in [...] Read more.
Proximity biotinylation, which utilizes various biotin ligating enzymes (BioID, TurboID, etc.), is widely used as a powerful tool for identifying novel protein–protein interactions. However, this method has a significant limitation: the use of streptavidin on beads for enriching biotinylated proteins often results in a high background of peptides from streptavidin itself, which interferes with identification by peptide mass fingerprinting. This limitation makes it practically impossible to study samples containing a small amount of material, such as individual insect tissues. In this study, we compared different precipitation and elution conditions for the purification of biotinylated proteins from protein extracts of Drosophila melanogaster S2 cells. We found that biotinylated proteins can be purified using anti-biotin antibodies, although with lower efficiency than streptavidin-based resin. We also demonstrated that protease-resistant streptavidin (prS), previously tested in mammalian cells, can be used effectively to purify biotinylated proteins from tissues of D. melanogaster. In our experiments, prS showed precipitation efficiency comparable to regular streptavidin but generated a lower background in peptide fingerprinting. To further demonstrate the applicability of prS for studying protein–protein interactions in D. melanogaster tissues, we carried out experiments to identify interaction partners of the ecdysone receptor (EcR) in D. melanogaster ovarian tissue using TurboID-based proximity biotinylation. As a result, EcR was found to interact with both previously described and novel protein partners in Drosophila ovaries. Full article
(This article belongs to the Special Issue Drosophila: A Versatile Model in Biology and Medicine—2nd Edition)
Show Figures

Figure 1

12 pages, 3454 KiB  
Article
DIRAS3 Inhibits Ovarian Cancer Cell Growth by Blocking the Fibronectin-Mediated Integrin β1/FAK/AKT Signaling Pathway
by Jing Guo, Janice M. Santiago-O’Farrill, Vivian Orellana, Rumeysa Ozyurt, Hailing Yang, Marc Pina, Gamze Bildik, Weiqun Mao, Robert C. Bast and Zhen Lu
Cells 2025, 14(16), 1250; https://doi.org/10.3390/cells14161250 - 13 Aug 2025
Viewed by 1203
Abstract
Autophagy is a crucial cellular process responsible for sustaining homeostasis through the degradation and recycling of proteins and organelles, providing energy during amino acid starvation and hypoxia. In cancer, autophagy can either inhibit tumor growth or support cancer cell survival. Our previous studies [...] Read more.
Autophagy is a crucial cellular process responsible for sustaining homeostasis through the degradation and recycling of proteins and organelles, providing energy during amino acid starvation and hypoxia. In cancer, autophagy can either inhibit tumor growth or support cancer cell survival. Our previous studies have shown that re-expression of the tumor suppressor gene DIRAS3 inhibits growth of ovarian cancer cells, promotes autophagic cell death in vitro, and induces tumor dormancy in vivo. Growth factors and extracellular matrix (ECM) components can, however, inhibit DIRAS3-induced autophagic cell death. This study explores whether fibronectin (FN) can counteract the growth inhibition induced by DIRAS3 in ovarian cancer cells. FN was found to inhibit DIRAS3-induced autophagy and to partially rescue ovarian cancer cells from DIRAS3-induced cell death while reducing DIRAS3-induced inhibition of p-FAK and p-AKT. Inhibiting FAK with defactinib in ovarian cancer cells enhanced DIRAS3-induced autophagy and cell death. Re-expression of DIRAS3 and treatment with defactinib produced tumor regression in xenograft models. Our findings suggest that ECM components in the tumor microenvironment like FN enhance the activities of β1 integrin, FAK, and AKT to inhibit DIRAS3-induced autophagic cell death, thereby promoting ovarian cancer cell survival. Full article
(This article belongs to the Section Autophagy)
Show Figures

Figure 1

16 pages, 1118 KiB  
Review
The Role of Receptor Tyrosine Kinase-like Orphan Receptor 1 (ROR1) in Cancer Stem Cell Signaling
by Matthew S. Jung, Won-Young Choi, Wenjing Zhang, Francisco N. Barrera and Rachel S. Perkins
Int. J. Mol. Sci. 2025, 26(16), 7828; https://doi.org/10.3390/ijms26167828 - 13 Aug 2025
Viewed by 377
Abstract
Receptor tyrosine kinase-like orphan receptor 1 (ROR1) is a key regulator of cancer stem cell (CSC) biology and signaling. In CSCs, ROR1 acts as a receptor or co-receptor, interacting with non-canonical WNT ligands, and forming complexes with proteins like CD19 and HER2, to [...] Read more.
Receptor tyrosine kinase-like orphan receptor 1 (ROR1) is a key regulator of cancer stem cell (CSC) biology and signaling. In CSCs, ROR1 acts as a receptor or co-receptor, interacting with non-canonical WNT ligands, and forming complexes with proteins like CD19 and HER2, to activate diverse downstream signaling pathways. ROR1 signaling in CSCs promotes proliferation, maintains stemness, and enhances migration, invasion, and the epithelial-to-mesenchymal transition (EMT). While minimally expressed after embryogenesis, ROR1 is aberrantly upregulated in numerous cancers, including ovarian, breast, pancreatic, and hematologic malignancies. ROR1 overexpression drives tumor progression, resistance to chemotherapies, disease recurrence, and ultimately metastasis. This expression pattern positions ROR1 as a promising target for CSC-specific therapies. High ROR1 expression is consistently linked to aggressive disease and poor patient outcomes. Here, we review ROR1′s role in CSCs and highlight the complex signaling that is observed in the CSC population. Further, we evaluate the gaps in the current understanding of ROR1 signaling in CSCs and describe how ROR1 regulates the associated signaling pathways. Finally, we provide an up-to-date summary of the promising therapeutic strategies targeting ROR1 that overcome conventional cancer treatment limitations. This review highlights the role of ROR1 as a critical, functional driver of CSCs and adverse patient outcomes across various malignancies. Full article
(This article belongs to the Special Issue New Advances in Cancer Stem Cell Research: 2nd Edition)
Show Figures

Figure 1

11 pages, 586 KiB  
Article
Fibroblast Activation Protein (FAP) as a Serum Biomarker for Fibrotic Ovarian Aging: A Clinical Validation Study Based on Translational Transcriptomic Targets
by Hyun Joo Lee, Yunju Jo, Shibo Wei, Eun Hee Yu, Sul Lee, Dongryeol Ryu and Jong Kil Joo
Int. J. Mol. Sci. 2025, 26(16), 7807; https://doi.org/10.3390/ijms26167807 - 13 Aug 2025
Viewed by 203
Abstract
Chronological age is an imprecise proxy for reproductive capacity, necessitating biomarkers that reflect the underlying pathophysiology of the ovary. Fibrotic remodeling of the ovarian stroma is a key hallmark of biological ovarian aging, yet it cannot be assessed by current clinical tools. This [...] Read more.
Chronological age is an imprecise proxy for reproductive capacity, necessitating biomarkers that reflect the underlying pathophysiology of the ovary. Fibrotic remodeling of the ovarian stroma is a key hallmark of biological ovarian aging, yet it cannot be assessed by current clinical tools. This study aimed to identify and validate a novel serum biomarker for fibrotic ovarian aging by applying supervised machine learning (ML) to human ovarian transcriptomic data. Transcriptomic data from the Genotype-Tissue Expression (GTEx) database were analyzed using ML algorithms to identify candidate genes predictive of ovarian aging, and finally, fibroblast activation protein (FAP) and collectin-11 (COLEC11) were selected for clinical validation. In a cross-sectional study, serum levels of FAP and COLEC11, along with key hormonal indices, were measured in two nested patient cohorts, and their associations with ovarian reserve and clinical parameters were analyzed. Serum FAP levels did not correlate with age but showed a strong inverse correlation with anti-Müllerian hormone (AMH) (r = −0.61, p = 0.001), a finding accentuated in women with decreased ovarian reserve (DOR). While COLEC11 correlated with age, it failed to differentiate DOR status. FAP levels were independent of central hormonal regulation, consistent with preclinical fibrotic models. Circulating FAP reflects age-independent, fibrotic ovarian aging, offering stromal-specific information not captured by conventional hormonal markers. This study provides the first clinical validation of FAP as a biomarker for ovarian stromal aging, holding potential for improved reproductive risk assessment. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

27 pages, 2676 KiB  
Review
Anticancer Activity of the Marine-Derived Compound Bryostatin 1: Preclinical and Clinical Evaluation
by Tomasz Kowalczyk, Marek Staszewski, Magdalena Markowicz-Piasecka, Joanna Sikora, Catarina Amaro, Laurent Picot and Przemysław Sitarek
Int. J. Mol. Sci. 2025, 26(16), 7765; https://doi.org/10.3390/ijms26167765 - 11 Aug 2025
Viewed by 327
Abstract
Bryostatin 1, a natural macrolide isolated from Bugula neritina, is a potent modulator of protein kinase C (PKC) isoforms with promising anticancer properties. In numerous in vitro studies, bryostatin 1 has been shown to inhibit tumor cell proliferation and induce differentiation and [...] Read more.
Bryostatin 1, a natural macrolide isolated from Bugula neritina, is a potent modulator of protein kinase C (PKC) isoforms with promising anticancer properties. In numerous in vitro studies, bryostatin 1 has been shown to inhibit tumor cell proliferation and induce differentiation and apoptotic cell death in a wide range of cell lines, including leukemia, lymphoma, glioma, and solid tumors such as ovarian and breast cancer. Its antitumor activity, both as monotherapy and in combination with conventional chemotherapy, has been confirmed in in vivo models, where synergistic effects have been observed, including sensitization of tumor cells to cytostatic agents. Despite promising preclinical findings, phase I and II clinical trials have not yielded the expected results, suggesting limited efficacy of the macrolide as a single agent with a relatively favorable safety profile. Current research directions focus on optimizing dosing regimens, combining bryostatin 1 with other anticancer drugs and identifying predictive biomarkers of response. This article reviews the current state of knowledge on the anticancer effects of bryostatin 1, analyzing available data from in vitro, in vivo, and clinical trials and discussing potential directions for further translational research. Full article
Show Figures

Figure 1

22 pages, 4751 KiB  
Article
Biophysical Insights into the Binding Interactions of Inhibitors (ICA-1S/1T) Targeting Protein Kinase C-ι
by Radwan Ebna Noor, Shahedul Islam, Tracess Smalley, Katarzyna Mizgalska, Mark Eschenfelder, Dimitra Keramisanou, Aaron Joshua Astalos, James William Leahy, Wayne Charles Guida, Aleksandra Karolak, Ioannis Gelis and Mildred Acevedo-Duncan
Biophysica 2025, 5(3), 36; https://doi.org/10.3390/biophysica5030036 - 11 Aug 2025
Viewed by 241
Abstract
The overexpression of atypical protein kinase C-iota (PKC-ι) is a biomarker for carcinogenesis in various cell types, such as glioma, ovarian, renal, etc., manifesting as a potential drug target. In previous in vitro studies, ICA-1S and ICA-1T, experimental candidates for inhibiting PKC-ι, have [...] Read more.
The overexpression of atypical protein kinase C-iota (PKC-ι) is a biomarker for carcinogenesis in various cell types, such as glioma, ovarian, renal, etc., manifesting as a potential drug target. In previous in vitro studies, ICA-1S and ICA-1T, experimental candidates for inhibiting PKC-ι, have demonstrated their specificity and promising efficacy against various cancers. Moreover, the in vivo studies have demonstrated low toxicity levels in acute and chronic murine models. Despite these prior developments, the binding affinities of the inhibitors were never thoroughly explored from a biophysical perspective. Here, we present the biophysical characterizations of PKC-ι in combination with ICA-1S/1T. Various methods based on molecular docking, light scattering, intrinsic fluorescence, thermal denaturation, and heat exchange were applied. The biophysical characteristics including particle sizing, thermal unfolding, aggregation profiles, enthalpy, entropy, free energy changes, and binding affinity (Kd) of the PKC-ι in the presence of ICA-1S were observed. The studies indicate the presence of domain-specific stabilities in the protein–ligand complex. Moreover, the results indicate a spontaneous reaction with an entropic gain, resulting in a possible entropy-driven hydrophobic interaction and hydrogen bonds in the binding pocket. Altogether, these biophysical studies reveal important insights into the binding interactions of PKC-ι and its inhibitors ICA-1S/1T. Full article
(This article belongs to the Collection Feature Papers in Biophysics)
Show Figures

Figure 1

14 pages, 2174 KiB  
Article
Effects of Diethylstilbestrol on Uterus Structure and Immunological Function in Mice During Early Pregnancy
by Jian Li, Ruiping Xu, Guan Wang, Yanhua Su, Yaoxing Chen and Jing Cao
Toxics 2025, 13(8), 672; https://doi.org/10.3390/toxics13080672 (registering DOI) - 9 Aug 2025
Viewed by 337
Abstract
Due to the growing environmental burden of endocrine-disrupting chemicals (EDCs), there is an increasing concern regarding the reproductive hazards posed by synthetic estrogens, particularly diethylstilbestrol (DES). However, the precise mechanisms by which DES disrupts uterine endocrine function and immune homeostasis leading to pregnancy [...] Read more.
Due to the growing environmental burden of endocrine-disrupting chemicals (EDCs), there is an increasing concern regarding the reproductive hazards posed by synthetic estrogens, particularly diethylstilbestrol (DES). However, the precise mechanisms by which DES disrupts uterine endocrine function and immune homeostasis leading to pregnancy failure remain unclear. Given that wild rodents serve as key reservoirs for zoonotic diseases such as plague, reproductive interventions targeting their pregnancy processes hold significant ecological implications for disease control. In this study, female mice in estrus were randomly divided into four experimental groups, receiving DES at doses of 0 (control), 0.02 (low), 0.2 (medium), and 2 mg/kg (high), respectively. For five consecutive days, mice were injected subcutaneously on a daily basis, with the goal of examining DES-related alterations in hormone secretion and local immune responses within the uterus and spleen. It was found that high-dose DES treatment significantly increased maternal body weight and spleen weight during early pregnancy (p < 0.05). Meanwhile, reproductive function declined progressively with increasing doses, as indicated by decreased ovarian and uterine weights, fewer embryos, and extended estrous cycle duration (p < 0.05). Hematoxylin and eosin staining revealed that high-dose DES markedly reduced uterine gland density at day P5, accompanied by epithelial vacuolar degeneration and nuclear pyknosis. The proportion of uterine glands relative to total uterine area also decreased significantly with increasing DES doses. Moreover, DES inhibited lymphocyte proliferation in both the uterus and spleen in a dose-dependent fashion, with ConA- and LPS-induced proliferation rates decreasing by 0.78–30.70% and 1.91–18.20%, respectively (p < 0.05). The proinflammatory cytokine IL-2 was significantly elevated by DES, whereas the anti-inflammatory cytokine IL-4 showed a notable decrease (p < 0.05). DES administration notably decreased uterine expression of proliferating cell nuclear antigen. In contrast, the numbers of B-cell lymphoma 2- and Bcl-2-associated X protein-positive cells rose, along with upregulated levels of inducible nitric oxide synthase. Furthermore, DES impaired antioxidant defenses in both the uterus and spleen, evidenced by the decreased activities of superoxide dismutase and glutathione peroxidase, reduced total antioxidant capacity, and elevated malondialdehyde levels. This study elucidates the multifaceted mechanisms by which DES impairs the early gestational reproductive environment, filling a critical knowledge gap regarding its interference with the uterus–immune axis, and expands the current understanding of the ecotoxicological impacts of endocrine-disrupting chemicals. Full article
(This article belongs to the Section Ecotoxicology)
Show Figures

Graphical abstract

15 pages, 4595 KiB  
Article
Oxidative Stress Triggers Porcine Ovarian Granulosa Cell Apoptosis Through MAPK Signaling
by Ting Zhao, Hui Jia, Xuerui Zhao, Xiaotong Gu, Chaoxiong Yong, Saihao Wang, Jiawei Zhou, Linrong Li, Mailin Gan, Lili Niu, Ye Zhao, Lei Chen, Xiaofeng Zhou, Linyuan Shen, Li Zhu and Yan Wang
Antioxidants 2025, 14(8), 978; https://doi.org/10.3390/antiox14080978 - 9 Aug 2025
Viewed by 389
Abstract
Follicle health determines the number and quality of sows’ ovulation, thereby influencing the litter size and the piglets’ viability. Granulosa cells (GCs) play a crucial role in follicular formation and development, and oxidative stress-induced GC death is a major cause of follicular dysplasia. [...] Read more.
Follicle health determines the number and quality of sows’ ovulation, thereby influencing the litter size and the piglets’ viability. Granulosa cells (GCs) play a crucial role in follicular formation and development, and oxidative stress-induced GC death is a major cause of follicular dysplasia. Previous studies have confirmed that oxidative stress triggers apoptosis in granulosa cells. In this study, we explored how oxidative stress influences apoptosis in porcine ovarian granulosa cells. We find that porcine atretic follicles exhibit significant oxidative stress, accompanied by the activation of the mitogen-activated protein kinase (MAPK) signaling pathway, including the upregulation of key factors such as apoptosis signal-regulating kinase 1 (ASK1). Healthy follicles of 3–5 mm were randomly assigned to the control group, H2O2 treatment group, and selonsertib pretreatment group. The porcine ovarian GCs were placed in cell culture medium supplemented with H2O2 to assess ROS production, cell proliferation, apoptosis, the expression levels of oxidative stress-related genes, and expression levels of apoptosis-related proteins. In vitro experiments in mouse GCs further confirmed that H2O2-induced oxidative stress triggers the upregulation of the MAPK pathway and promotes granulosa cell apoptosis. The results showed that H2O2 treatment induced ROS production and apoptosis in porcine GCs and inhibited GC viability. Additionally, selonsertib pretreatment attenuated apoptosis in GCs by inhibiting H2O2-induced oxidative stress. In summary, our findings reveal that oxidative stress induced granulosa cell apoptosis via the MAPK signaling pathway, impairing proper follicular development in pigs. Full article
Show Figures

Figure 1

15 pages, 3491 KiB  
Article
PARP Inhibition Shifts Murine Myeloid Cells Toward a More Tolerogenic Profile In Vivo
by Jose R. Pittaluga-Villarreal, Casey M. Daniels, Tara Capece, Pauline R. Kaplan, Martin Meier-Schellersheim and Aleksandra Nita-Lazar
Biomolecules 2025, 15(8), 1149; https://doi.org/10.3390/biom15081149 - 9 Aug 2025
Viewed by 446
Abstract
The human Poly ADP-ribose Polymerase (PARP) family comprises 17 enzymes responsible for the transfer of ADP-ribose to proteins, forming poly- or mono-ADP-ribosylation. This post-translational modification regulates DNA repair and programmed cell death, processes affecting cancer biology. PARP inhibitors, including the FDA-approved olaparib, are [...] Read more.
The human Poly ADP-ribose Polymerase (PARP) family comprises 17 enzymes responsible for the transfer of ADP-ribose to proteins, forming poly- or mono-ADP-ribosylation. This post-translational modification regulates DNA repair and programmed cell death, processes affecting cancer biology. PARP inhibitors, including the FDA-approved olaparib, are used to treat BRCA-dependent breast and ovarian cancers. Although therapies with use of PARP inhibitors are showing clinical success, their effects on the immune system remain understudied. Prior work has shown that PARP inhibition can modulate inflammatory responses and alter innate immunity. In this study, we evaluated the immunomodulatory effects of olaparib on myeloid cells in vivo, focusing on bone marrow and spleen. Olaparib treatment altered the composition and activation state of dendritic cells, neutrophils, and macrophages. In the bone marrow, olaparib increased the proportion of cDC2 population, mature neutrophils and inflammatory macrophages expressing CD80. In contrast, splenic myeloid cells exhibited enhanced expression of markers associated with tolerogenic phenotypes, including CD206 and CD124 in neutrophils and macrophages. The spleen also showed an increase in immature monocyte-derived dendritic cells (CD206+) and a bias toward the cDC2 subset. These findings indicate that PARP inhibition can induce short-term phenotypic remodeling of myeloid cell populations, promoting a more immunoregulatory profile, especially in the spleen. These changes may contribute to an altered immune landscape with implications for anti-tumor immunity. Full article
(This article belongs to the Special Issue PARPs in Cell Death and PARP Inhibitors in Cancers: 2nd Edition)
Show Figures

Figure 1

18 pages, 2516 KiB  
Article
Joint Metabolomics and Transcriptomics Reveal Rewired Glycerophospholipid and Arginine Metabolism as Components of BRCA1-Induced Metabolic Reprogramming in Breast Cancer Cells
by Thomas Lucaora and Daniel Morvan
Metabolites 2025, 15(8), 534; https://doi.org/10.3390/metabo15080534 - 7 Aug 2025
Viewed by 398
Abstract
Background/Objectives: The breast cancer susceptibility gene 1 (BRCA1) is a tumor suppressor gene whose mutations are associated with increased susceptibility to develop breast or ovarian cancer. BRCA1 mainly exerts its protective effects through DNA double-strand break repair. Although not itself [...] Read more.
Background/Objectives: The breast cancer susceptibility gene 1 (BRCA1) is a tumor suppressor gene whose mutations are associated with increased susceptibility to develop breast or ovarian cancer. BRCA1 mainly exerts its protective effects through DNA double-strand break repair. Although not itself a transcriptional factor, BRCA1, through its multiple protein interaction domains, exerts transcriptional coregulation. In addition, BRCA1 expression alters cellular metabolism including inhibition of de novo fatty acid synthesis, changes in cellular bioenergetics, and activation of antioxidant defenses. Some of these actions may contribute to its global oncosuppressive effects. However, the breadth of metabolic pathways reprogrammed by BRCA1 is not fully elucidated. Methods: Breast cancer cells expressing BRCA1 were investigated by multiplatform metabolomics, metabolism-related transcriptomics, and joint metabolomics/transcriptomics data processing techniques, namely two-way orthogonal partial least squares and pathway analysis. Results: Joint analyses revealed the most important metabolites, genes, and pathways of metabolic reprogramming in BRCA1-expressing breast cancer cells. The breadth of metabolic reprogramming included fatty acid synthesis, bioenergetics, HIF-1 signaling pathway, antioxidation, nucleic acid synthesis, and other pathways. Among them, rewiring of glycerophospholipid (including phosphatidylcholine, -serine and -inositol) metabolism and increased arginine metabolism have not been reported yet. Conclusions: Rewired glycerophospholipid and arginine metabolism were identified as components of BRCA1-induced metabolic reprogramming in breast cancer cells. The study helps to identify metabolites that are candidate biomarkers of the BRCA1 genotype and metabolic pathways that can be exploited in targeted therapies. Full article
(This article belongs to the Section Cell Metabolism)
Show Figures

Figure 1

16 pages, 2369 KiB  
Article
HMGB1 Deficiency Occurs in a Broad Range of Human Cancers and Is Often Associated with Unfavorable Tumor Phenotype
by Viktoria Chirico, Hena Sharifi, Maria Christina Tsourlakis, Seyma Büyücek, Clara Marie von Bargen, Katharina Möller, Florian Lutz, David Dum, Martina Kluth, Claudia Hube-Magg, Georgia Makrypidi-Fraune, Piero Caneve, Maximilian Lennartz, Morton Freytag, Sebastian Dwertmann Rico, Simon Kind, Viktor Reiswich, Eike Burandt, Till S. Clauditz, Patrick Lebok, Christoph Fraune, Till Krech, Sarah Minner, Andreas H. Marx, Waldemar Wilczak, Ronald Simon, Guido Sauter, Stefan Steurer and Kristina Jansenadd Show full author list remove Hide full author list
Diagnostics 2025, 15(15), 1974; https://doi.org/10.3390/diagnostics15151974 - 6 Aug 2025
Viewed by 367
Abstract
Background/Objectives: Aberrant expression of high-mobility group protein B1 (HMGB1) has been linked to cancer development and progression. Methods: To better comprehend the role of HMGB1 expression in cancer, a tissue microarray containing 14,966 samples from 134 different tumor entities and 608 [...] Read more.
Background/Objectives: Aberrant expression of high-mobility group protein B1 (HMGB1) has been linked to cancer development and progression. Methods: To better comprehend the role of HMGB1 expression in cancer, a tissue microarray containing 14,966 samples from 134 different tumor entities and 608 samples of 76 different normal tissue types was analyzed by immunohistochemistry. Results: Strong HMGB1 staining occurred in almost all normal cell types and in most cancers. Of 11,808 evaluable cancers, only 7.8% showed complete absence of HMGB1 staining (HMGB1 deficiency) while 9.9% showed 1+, 25.0% showed 2+, and 57.2% showed 3+ HMGB1 positivity. Absence of HMGB1 staining mostly occurred in pheochromocytoma (90.0%), seminoma (72.4%), gastrointestinal stromal tumor (28.6%), adrenal cortical carcinoma (25.0%), and Hodgkin’s lymphoma (25.0%). Low HMGB1 staining was linked to poor histologic grade (p < 0.0001), advanced pT stage (p < 0.0001), high UICC stage (p < 0.0001), and distant metastasis (p = 0.0413) in clear cell renal cell carcinoma, invasive tumor growth in urothelial carcinoma (pTa vs. pT2–4, p < 0.0001), mismatch repair deficiency (p = 0.0167) in colorectal cancers, and advanced pT stage in invasive breast carcinoma of no special type (p = 0.0038). Strong HMGB1 staining was linked to nodal metastases in high-grade serous ovarian carcinomas (p = 0.0213) and colorectal adenocarcinomas (p = 0.0137), as well as to poor histological grade in squamous cell carcinomas (p = 0.0010). Conclusions: HMGB1 deficiency and reduced HMGB1 expression occur in a broad range of different tumor entities. Low rather than strong HMGB1 staining is often linked to an aggressive tumor phenotype. Whether HMGB1 deficiency renders cells susceptible to specific drugs remains to be determined. Full article
(This article belongs to the Section Pathology and Molecular Diagnostics)
Show Figures

Figure 1

18 pages, 5256 KiB  
Article
Impact of Alginate Oligosaccharides on Ovarian Performance and the Gut Microbial Community in Mice with D-Galactose-Induced Premature Ovarian Insufficiency
by Yan Zhang, Hongda Pan, Dao Xiang, Hexuan Qu and Shuang Liang
Antioxidants 2025, 14(8), 962; https://doi.org/10.3390/antiox14080962 - 5 Aug 2025
Viewed by 388
Abstract
Premature ovarian insufficiency (POI) is an important factor in female infertility and is often associated with oxidative stress. Alginate oligosaccharides (AOSs), derived from the degradation of alginate, have been demonstrated to have protective effects against various oxidative stress-related diseases. However, the impact of [...] Read more.
Premature ovarian insufficiency (POI) is an important factor in female infertility and is often associated with oxidative stress. Alginate oligosaccharides (AOSs), derived from the degradation of alginate, have been demonstrated to have protective effects against various oxidative stress-related diseases. However, the impact of AOSs on POI has not been previously explored. The current study explored the effects of AOSs on ovarian dysfunction in a mouse model of POI induced by D-galactose (D-gal). Female C57BL/6 mice were randomly divided into five groups: the control (CON), POI model (D-gal), and low-, medium-, and high-dose AOS groups (AOS-L, 100 mg/kg/day; AOS-M, 150 mg/kg/day; AOS-H, 200 mg/kg/day). For 42 consecutive days, mice in the D-gal, AOS-L, AOS-M, and AOS-H groups received daily intraperitoneal injections of D-gal (200 mg/kg/day), whereas those in the CON group received equivalent volumes of sterile saline. Following D-gal injection, AOSs were administered via gavage at the specified doses; mice in the CON and D-gal groups received sterile saline instead. AOS treatment markedly improved estrous cycle irregularities, normalized serum hormone levels, reduced granulosa cell apoptosis, and increased follicle counts in POI mice. Moreover, AOSs significantly reduced ovarian oxidative stress and senescence in POI mice, as indicated by lower levels of malondialdehyde (MDA), higher activities of catalase (CAT) and superoxide dismutase (SOD), and decreased protein expression of 4-hydroxynonenal (4-HNE), nitrotyrosine (NTY), 8-hydroxydeoxyguanosine (8-OHdG), and p16 in ovarian tissue. Analysis of the gut microbiota through 16S rRNA gene sequencing and short-chain fatty acid (SCFA) analysis revealed significant differences in gut microbiota composition and SCFA levels (acetic acid and total SCFAs) between control and D-gal-induced POI mice. These differences were largely alleviated by AOS treatment. AOSs changed the gut microbiota by increasing the abundance of Ligilactobacillus and decreasing the abundance of Clostridiales, Clostridiaceae, Marinifilaceae, and Clostridium_T. Additionally, AOSs mitigated the decline in acetic acid and total SCFA levels observed in POI mice. Notably, the total SCFA level was significantly correlated with the abundance of Ligilactobacillus, Marinifilaceae, and Clostridium_T. In conclusion, AOS intervention effectively mitigates ovarian oxidative stress, restores gut microbiota homeostasis, and regulates the microbiota–SCFA axis, collectively improving D-gal-induced POI. Therefore, AOSs represent a promising therapeutic strategy for POI management. Full article
(This article belongs to the Section Health Outcomes of Antioxidants and Oxidative Stress)
Show Figures

Figure 1

Back to TopTop