251
|
Qi Y, Lin C, Zhao C, Wu Y. Circ_0003072 Mediates the Pro-osteogenic Differentiation Effect of Betulinic Acid on Human Periodontal Ligament Stem Cells. Int Dent J 2025; 75:1390-1399. [PMID: 39572284 PMCID: PMC11976600 DOI: 10.1016/j.identj.2024.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/13/2024] [Accepted: 10/27/2024] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Betulinic acid (BetA) exhibits a good pro-osteogenic differentiation effect on human periodontal ligament stem cells (hPDLSCs), making it a promising supplement for periodontal regeneration. Circular RNAs (circRNAs) have emerged as important regulators of cellular behaviour, and whether circRNAs are involved in the effects of BetA remains unknown. METHODS Bioinformatics analysis was used to screen for dysregulated circRNAs involved in osteogenic differentiation based on public datasets. Osteogenic differentiation was evaluated using quantitative PCR detection of RUNX2, ALPL, COL1A1, and BGLAP levels, alkaline phosphatase staining, and Alizarin Red S staining of calcified nodules. The role of circ_0003072 in the BetA-regulated osteogenic differentiation of hPDLSCs was investigated, and its downstream microRNAs and mRNA were confirmed using RNA-sequencing, competing endogenous RNA network construction, gene ontology analysis, dual-luciferase reporter assays, and functional assays. RESULTS circ_0003072 showed the highest fold-change among the 18 candidate circRNAs, and knockdown of circ_0003072 inhibited the pro-osteogenic differentiation effect of BetA on hPDLSCs. RNA-sequencing combined with gene ontology analysis identified 11 osteogenesis-relevant genes. Five genes that shared microRNAs between circ_0003072 and two candidate genes (chordin-like 1 [CHRDL1] and XIAP) were screened, and miR-488-3p showed the highest increase after silencing circ_0003072. Knockdown of circ_0003072 inhibited the pro-osteogenic differentiation effect of silencing miR-488-3p. miR-488-3p bound to circ_0003072 and CHRDL1. Furthermore, overexpression of CHRDL1 rescued the miR-488-3p-induced inhibition of osteogenic differentiation. CONCLUSION BetA promoted the osteogenic differentiation of hPDLSCs by regulating the circ_0003072/miR-488-3p/CHRDL1 pathway, and circ_0003072 acted as a sponge for miR-488-3p, thus upregulating the level of CHRDL1.
Collapse
Affiliation(s)
- Yuesun Qi
- Department of Stomatology, Jinshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Chunnan Lin
- Department of Stomatology, Jinshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Chengwei Zhao
- Department of Stomatology, Jinshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Ye Wu
- Department of Stomatology, Fengxian District Institute of Dental Diseases, Shanghai, People's Republic of China.
| |
Collapse
|
252
|
Mu X, Feng L, Wang Q, Li H, Zhou H, Yi W, Sun Y. Decreased gut microbiome-derived indole-3-propionic acid mediates the exacerbation of myocardial ischemia/reperfusion injury following depression via the brain-gut-heart axis. Redox Biol 2025; 81:103580. [PMID: 40058066 PMCID: PMC11930714 DOI: 10.1016/j.redox.2025.103580] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 02/11/2025] [Accepted: 02/19/2025] [Indexed: 03/22/2025] Open
Abstract
Despite the increasing recognition of the interplay between depression and cardiovascular disease (CVD), the precise mechanisms by which depression contributes to the pathogenesis of cardiovascular disease remain inadequately understood. The involvement of gut microbiota and their metabolites to health and disease susceptibility has been gaining increasing attention. In this study, it was found that depression exacerbated cardiac injury, impaired cardiac function (EF%: P < 0.01; FS%: P < 0.05), hindered long-term survival (P < 0.01), and intensified adverse cardiac remodeling (WGA: P < 0.01; MASSON: P < 0.0001) after myocardial ischemia/reperfusion (MI/R) in mice. Then we found that mice receiving microbiota transplants from chronic social defeat stress (CSDS) mice exhibited worse cardiac function (EF%: P < 0.01; FS%: P < 0.01) than those receiving microbiota transplants from non-CSDS mice after MI/R injury. Moreover, impaired tryptophan metabolism due to alterations in gut microbiota composition and structure was observed in the CSDS mice. Mechanistically, we analyzed the metabolomics of fecal and serum samples from CSDS mice and identified indole-3-propionic acid (IPA) as a protective agent for cardiomyocytes against ferroptosis after MI/R via NRF2/System xc-/GPX4 axis, played a role in mediating the detrimental influence of depression on MI/R. Our findings provide new insights into the role of the gut microbiota and IPA in depression and CVD, forming the basis of intervention strategies aimed at mitigating the deterioration of cardiac function following MI/R in patients experiencing depression.
Collapse
Affiliation(s)
- Xingdou Mu
- Department of Geriatric, XiJing Hospital, Xi'an, Shaanxi, 710000, China
| | - Lele Feng
- Department of Cardiovascular Surgery, XiJing Hospital, Xi'an, Shaanxi, 710000, China
| | - Qiang Wang
- Department of Geriatric, XiJing Hospital, Xi'an, Shaanxi, 710000, China
| | - Hong Li
- Department of Geriatric, XiJing Hospital, Xi'an, Shaanxi, 710000, China
| | - Haitao Zhou
- Department of Geriatric, XiJing Hospital, Xi'an, Shaanxi, 710000, China
| | - Wei Yi
- Department of Cardiovascular Surgery, XiJing Hospital, Xi'an, Shaanxi, 710000, China.
| | - Yang Sun
- Department of Geriatric, XiJing Hospital, Xi'an, Shaanxi, 710000, China.
| |
Collapse
|
253
|
Metri NA, Mandl A, Paller CJ. Harnessing nature's therapeutic potential: A review of natural products in prostate cancer management. Urol Oncol 2025; 43:221-243. [PMID: 39794185 DOI: 10.1016/j.urolonc.2024.12.260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 11/16/2024] [Accepted: 12/01/2024] [Indexed: 01/13/2025]
Abstract
Prostate cancer is the most commonly diagnosed cancer and the second leading cause of cancer-related death among men in the United States. The global burden of this disease is rising, placing significant strain on healthcare systems worldwide. Although definitive therapies like surgery and radiation are often effective, prostate cancer can recur and progress to castration-resistant prostate cancer in some cases. Conventional treatments for prostate cancer often have substantial side effects that can greatly impact patients' quality of life. Therefore, many patients turn to complementary therapies to improve outcomes, manage side effects, and enhance overall well-being. Natural products show promise as complementary treatments for prostate cancer, offering anticancer properties with a low risk of adverse effects. While preclinical research has produced encouraging results, their role in prostate cancer treatment remains controversial, largely due to inconsistent and limited success in clinical trials. This review explores the mechanisms of action of key natural products in prostate cancer management and summarizes clinical trials evaluating their efficacy and safety. It underscores the need for high-quality, rigorously designed, and adequately powered studies to validate the therapeutic potential and safety of these supplements in cancer care. Additionally, we propose future directions to enhance their role in addressing the complex challenges associated with prostate cancer.
Collapse
Affiliation(s)
- Nicole A Metri
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD
| | - Adel Mandl
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD
| | - Channing J Paller
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD.
| |
Collapse
|
254
|
Hu M, Zheng L, Li A, Li X, Liang W, Zhu Y, Wang A, He L, Liu X, Sun Q. Discovery of 3-indolylbenzoquinone derivatives with therapeutic potential for breast cancer. Bioorg Med Chem 2025; 120:118094. [PMID: 39933277 DOI: 10.1016/j.bmc.2025.118094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 01/25/2025] [Accepted: 01/29/2025] [Indexed: 02/13/2025]
Abstract
Breast cancer is one of the most prevalent malignant tumors in women, but the side effects and drug resistance limit the long-term effectiveness of existing drugs. To address these issues, we designed and synthesized a series of novel mono- and bis-indole-substituted 3-indolylbenzoquinone derivatives and evaluated their inhibitory activity against breast cancer. Among them, compound 1b demonstrated the most potent inhibitory activity against the MDA-MB-231 breast cancer cell line (IC50 = 3.2 µM) as well as the drug-resistant variant, MCF-7/ADR (IC50 = 8.36 µM). It demonstrated minimal toxicity and superior tumor suppression in a Balb/c mouse model of 4 T1 breast cancer. Mechanistically, compound 1b induced apoptosis and cell cycle arrest at the G2/M phase. Through computational study and CESTA assay, we implicated phosphoinositide 3-kinase α (PI3Kα) as a potential target. Thus, we present compound 1b as a lead candidate for the development of novel, safe, and effective small-molecule therapies against breast cancer.
Collapse
Affiliation(s)
- Mingli Hu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Lang Zheng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Ailing Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Xiao Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Wengxue Liang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Yuanhao Zhu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Aoxue Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Ling He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Xiuxiu Liu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, PR China.
| | - Qiu Sun
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
255
|
Alhalmi A, Amin S, Ralli T, Ali KS, Kohli K. Therapeutic role of naringin in cancer: molecular pathways, synergy with other agents, and nanocarrier innovations. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:3595-3615. [PMID: 39614898 DOI: 10.1007/s00210-024-03672-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 11/24/2024] [Indexed: 04/10/2025]
Abstract
Naringin, a flavanone glycoside found abundantly in citrus fruits, is well-known for its various pharmacological properties, particularly its significant anticancer effects. Research, both in vitro and in vivo, has shown that naringin is effective against several types of cancer, including liver, breast, thyroid, prostate, colon, bladder, cervical, lung, ovarian, brain, melanoma, and leukemia. Its anticancer properties are mediated through multiple mechanisms, such as apoptosis induction, inhibition of cell proliferation, cell cycle arrest, and suppression of angiogenesis, metastasis, and invasion, all while exhibiting minimal toxicity and adverse effects. Naringin's molecular mechanisms involve the modulation of essential signaling pathways, including PI3K/Akt/mTOR, FAK/MMPs, FAK/bads, FAKp-Try397, IKKs/IB/NF-κB, JNK, ERK, β-catenin, p21CIPI/WAFI, and p38-MAPK. Additionally, it targets several signaling proteins, such as Bax, TNF-α, Zeb1, Bcl-2, caspases, VEGF, COX-2, VCAM-1, and interleukins, contributing to its wide-ranging antitumor effects. The remarkable therapeutic potential of naringin, along with its favorable safety profile, highlights its promise as a candidate for cancer treatment. This comprehensive review examines the molecular mechanisms behind naringin's chemopreventive and anticancer effects, including its pharmacokinetics and bioavailability. Furthermore, it discusses advancements in nanocarrier technologies designed to enhance these characteristics and explores the synergistic benefits of combining naringin with other anticancer agents, focusing on improved therapeutic efficacy and drug bioavailability.
Collapse
Affiliation(s)
- Abdulsalam Alhalmi
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
- Department of Pharmaceutics, Faculty of Pharmacy, University of Aden, Aden, Yemen
| | - Saima Amin
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Tanya Ralli
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
- School of Pharmacy, COER University, Roorkee, 247667, India
| | - Khaled Saeed Ali
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Aden, Aden, Yemen
| | - Kanchan Kohli
- Faculty of Pharmacy, Lloyd Institute of Management and Technology, Greater Noida, 201308, India.
- Department of Pharmaceutical Sciences, Gurugram University, Haryana, 122003, India.
| |
Collapse
|
256
|
EFSA Panel on Additives and Products or Substances used in Animal Feed (FEEDAP), Villa RE, Azimonti G, Bonos E, Christensen H, Durjava M, Dusemund B, Gehring R, Glandorf B, Kouba M, López‐Alonso M, Marcon F, Nebbia C, Pechová A, Prieto‐Maradona M, Röhe I, Theodoridou K, Bampidis V, Dierick N, Woutersen R, de Lourdes Bastos M, Ramos F, Galobart J, Vettori MV, Holczknecht O, Ortuño J, Pizzo F, Valeri P, Innocenti ML, Amaduzzi A. Safety and efficacy of a feed additive consisting of Kieselguhr (diatomaceous earth) for all animal species (Imerys France). EFSA J 2025; 23:e9363. [PMID: 40242779 PMCID: PMC12000819 DOI: 10.2903/j.efsa.2025.9363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025] Open
Abstract
Following a request from the European Commission, EFSA was asked to deliver a scientific opinion on the safety and efficacy of Kieselguhr (diatomaceous earth, three forms: natural, purified calcined and purified flux-calcined) as a technological additive (functional group: anticaking and binder) for all animal species. Kieselguhr is predominantly amorphous silica (non-crystalline silicon dioxide (SiO2)). Based on the information submitted, Kieselguhr natural, purified calcined and purified flux-calcined are safe when used at 5000 mg/kg of complete feed in all terrestrial animals. No conclusions can be reached on the safety of its use in aquatic animals. The use of Kieselguhr natural, purified calcined and purified flux-calcined in animal nutrition under the proposed conditions of use is safe for the consumer and the environment. Kieselguhr natural and purified flux-calcined are not irritant to the skin. All the three forms of the additive are irritant to the eyes and should be considered dermal and respiratory sensitisers. Any exposure is considered a risk. The FEEDAP Panel concluded that the additive is effective as a pellet binder. Kieselguhr is efficacious as an anticaking agent at a minimum concentration of 15,000 mg/kg.
Collapse
|
257
|
Zhang J, Wang Z, Zhang R, Lei X, Wang G, Zou P. Hemicyanine-Phenothiazine Based Highly Selective Ratiometric Fluorescent Probes for Detecting Hypochlorite Ion in Fruits, Vegetables and Beverages. J Fluoresc 2025; 35:2449-2459. [PMID: 38607530 DOI: 10.1007/s10895-024-03694-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 03/26/2024] [Indexed: 04/13/2024]
Abstract
Hypochloric acid (HClO) is a reactive oxygen species (ROS) that functions as a bacteriostatic and disinfectant in food production. Excessive levels of ClO-, however, have been linked to various health issues, including cardiovascular diseases (Halliwell and Gutteridge in Oxford University press, USA, 2015), arthritis, and neurodegenerative diseases (Heinzelmann and Bauer in Biol Chem. 391(6):675-693, 2010). Therefore, synthesizing highly selective and sensitive probes for rapidly detecting endogenous ClO- in daily foods is currently a popular research topic (Kalyanaraman et al. in Redox Biol. 15:347-362, 2018; Winterbourn in Nat Chem Biol. 4(5):278-286, 2008; Turrens in J Physiol. 552(2):335-344, 2003). Thus, we have developed two highly selective ratiometric fluorescent probes (Probe1 and Probe2) based on indole-phenothiazine to detect ClO- in common vegetables, fruits and beverages qualitatively and quantitatively. Moreover, Both Probe1 and Probe2 have shown good specificity and stability, with high fluorescence intensity and long duration (Feng et al. in Adv Sci. 5:1800397, 2018; Wei et al. in Angew Chem. 131(14):4595-4599, 2019; Baruah et al. in J Mater Chem B, 2022).
Collapse
Affiliation(s)
- Jinyang Zhang
- College of Science, Sichuan Agricultural University, Ya'an, 625014, People's Republic of China
| | - Zhe Wang
- College of Science, Sichuan Agricultural University, Ya'an, 625014, People's Republic of China
| | - Rui Zhang
- College of Science, Sichuan Agricultural University, Ya'an, 625014, People's Republic of China
| | - Xueli Lei
- College of Science, Sichuan Agricultural University, Ya'an, 625014, People's Republic of China
| | - Guangtu Wang
- College of Science, Sichuan Agricultural University, Ya'an, 625014, People's Republic of China.
| | - Ping Zou
- College of Science, Sichuan Agricultural University, Ya'an, 625014, People's Republic of China.
| |
Collapse
|
258
|
Peng G, Pan X, Ye Z, Yi X, Xie Q, Zhang X, Tong N. Nongenetic risk factors for thyroid cancer: an umbrella review of evidence. Endocrine 2025; 88:60-74. [PMID: 39745600 DOI: 10.1007/s12020-024-04155-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 12/27/2024] [Indexed: 01/12/2025]
Abstract
BACKGROUND The incidence of thyroid cancer has increased annually, but the risk factors for thyroid cancer are still unclear. In this umbrella review, we aimed to identify associations between nongenetic risk factors and thyroid cancer incidence, and assess the quality and validity of the evidence. METHODS PubMed, Embase and the Cochrane Database of Systematic Reviews were searched to identify related meta-analyses or systematic reviews of epidemiological studies. We extracted the estimated summary effect and 95% confidence interval (CI) through fixed or random effects models of each meta-analysis. AMSTAR2 and the Grading of Recommendations, Assessment, Development and Evaluation (GRADE) were used to evaluate the methodological quality of the included meta-analyses and the quality of evidence respectively. Further subgroup analyses by sex and sensitivity analyses were conducted. RESULTS We identified 53 articles with 112 associations, of which 69 had significant relationships with thyroid cancer risk, including factors related to iodine, nitrates, fish, vitamin D, tea, alcohol, smoke, body mass index (BMI), pesticides, X-ray, I131, oral contraceptives, flavonoids, reproductive factors and some medical conditions. However, most studies (65%) were categorized as "critically low" on the basis of AMSTAR2, and most evidence (86%) was of weak quality since the classification by GRADE was very low. Moreover, subgroup and sensitivity analyses revealed more risk factors in women than in men. CONCLUSION We found that several modifiable factors have essential effects in the primary prevention of thyroid cancer, but few high-quality studies exist. In the future, more well-conducted, especially prospective, studies are needed to confirm the results. TRIAL REGISTRATION The protocol for this review was registered in PROSPERO (CRD42022352841).
Collapse
Affiliation(s)
- Ge Peng
- Division of Endocrinology and Metabolism, Laboratory of Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaohui Pan
- Division of Endocrinology and Metabolism, Laboratory of Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
| | - Ziwei Ye
- Division of Endocrinology and Metabolism, Laboratory of Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
| | - Xianyanling Yi
- Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Qingxing Xie
- Division of Endocrinology and Metabolism, Laboratory of Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
| | - Xinyi Zhang
- Division of Endocrinology and Metabolism, Laboratory of Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
| | - Nanwei Tong
- Division of Endocrinology and Metabolism, Laboratory of Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
259
|
Montoya G, van Ravenzwaay B, Seefelder W, Haake V, Kamp H. Unanticipated differences in the rat plasma metabolome of genistein and daidzein. Arch Toxicol 2025; 99:1387-1406. [PMID: 39954026 PMCID: PMC11968494 DOI: 10.1007/s00204-025-03967-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/15/2025] [Indexed: 02/17/2025]
Abstract
Genistein (GEN) and daidzein (DAI) are soy isoflavones known to bind to estrogen receptors. Overall health effects of GEN and DAI in humans exhibit a dual nature, presenting both health benefits and concerns related to their interaction with the estrogen receptor. The metabolomes of these isoflavones were determined in 28-day oral studies in male and female Wistar rats to elucidate (1) metabolites changes, (2) compare their metabolomes with other compounds and (3) identify toxicological modes of action (MoA). Dose levels for GEN were 1000 and 300 mg/kg bw by gavage and 1000 and 300 ppm (via diet). DAI gavage dose levels were 1000 and 100 mg/kg bw. Results were evaluated using the MetaMap®Tox data base. Both compounds demonstrated metabolome profiles which were associated with estrogenic profiles and compounds, predominantly in females. However, the metabolomes were compound specific with relatively few common metabolite changes. There were no relevant matches between any GEN and any DAI treatment group indicating that both compounds are substantially different from metabolome perspective. Ranking of the metabolome patters for GEN and DAI with ≥ 1000 compounds in the MetaMap®Tox database revealed correlations with estrogenic and other hormonally active compounds. GEN-treated females correlated best with Cabergoline, a dopamine D2 receptor agonist, DAI females with tamoxifen and diethylstilbestrol, suggesting that even their estrogenic activity may be different. Beyond estrogenic effects, the high dose (HD) DAI metabolome indicated altered fatty acid metabolism associated with PPAR-alpha activation. For GEN, there was an indication of ethanolamine-like liver effects. Dose levels without estrogenic effects for GEN were 1000 and 100 mg/kg bw for males and females respectively, there were no estrogenic effects in the feeding studies. For DAI males, the no estrogenic effect level was 300 mg/kg bw, for females < 100 mg/kg bw, suggesting that DAI may be a more potent estrogen than GEN in rats.
Collapse
Affiliation(s)
- Gina Montoya
- Société Des Produits Nestlé S.A, Lausanne, Switzerland
| | | | | | | | | |
Collapse
|
260
|
Jiang C, Zhu Y, Zhang J, Chen H, Li W, Xie R, Kong L, Chen L, Chen X, Huang H, Xu S. NR4A1 suppresses breast cancer growth by repressing c-Fos-mediated lipid and redox dyshomeostasis. Exp Mol Med 2025; 57:804-819. [PMID: 40164686 PMCID: PMC12045962 DOI: 10.1038/s12276-025-01430-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 12/31/2024] [Accepted: 01/13/2025] [Indexed: 04/02/2025] Open
Abstract
The specific function of NR4A1 as a transcriptional regulator in cancer remains unclear. Here we report the biological effect of NR4A1 in suppressing breast cancer (BC) growth. We found that NR4A1 deficiency was correlated with BC progression in the clinic. Genetic deletion of NR4A1 in BC cells significantly promoted cellular proliferation and tumor growth. Moreover, global metabolome screening indicated that the deletion of NR4A1 resulted in tumor lipid remodeling and phospholipid accumulation, which was accompanied by increases in fatty acid and lipid uptake. In addition, NR4A1 knockout induced oxidative stress that aggravated redox balance disruption. Mechanistically, transcriptomic and epigenomic analyses revealed that NR4A1 restrained BC cell proliferation by directly interacting with c-Fos and competitively inhibiting c-Fos binding to the promoter of the target gene PRDX6, which is involved in lipid and redox homeostasis. Notably, we confirmed that the treatment of BC cells with the selective NR4A1 agonist cytosporone B significantly activated the expression of NR4A1, followed by increased interaction between NR4A1 and c-Fos, thereby interfering with c-Fos-mediated transcriptional regulation of BC cell growth. Thus, NR4A1 plays a vital role in reducing the c-Fos-induced activation of downstream signaling cascades in BC, suggesting that agents that activate NR4A1 may be potential therapeutic strategies.
Collapse
Affiliation(s)
- Cen Jiang
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China.
| | - Youzhi Zhu
- Department of Thyroid and Breast Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Thyroid and Breast Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Junsi Zhang
- Department of Thyroid and Breast Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Huaying Chen
- Department of Thyroid and Breast Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Weiwei Li
- Department of Thyroid and Breast Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Ruiwang Xie
- Department of Thyroid and Breast Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Lingjun Kong
- Department of Thyroid and Breast Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Thyroid and Breast Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Ling Chen
- Department of Thyroid and Breast Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Thyroid and Breast Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Xiangjin Chen
- Department of Thyroid and Breast Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Thyroid and Breast Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Huifang Huang
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China.
| | - Sunwang Xu
- Department of Thyroid and Breast Surgery, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
- Department of Thyroid and Breast Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
- Fujian Provincial Key Laboratory of Precision Medicine for Cancer, Fuzhou, China.
| |
Collapse
|
261
|
K N S, Shetty P, Devaranagadi B, Hundekari IA. Targeting ERβ1-Positive Triple-Negative Breast Cancer: Molecular Effects of Calcitriol and 17β-Estradiol. Cureus 2025; 17:e82934. [PMID: 40416168 PMCID: PMC12103257 DOI: 10.7759/cureus.82934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 04/14/2025] [Indexed: 05/27/2025] Open
Abstract
Background Breast cancer is the most common malignancy in women. Triple-negative breast cancer (TNBC) is characterized by the absence of estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2, contributing to its aggressive nature, limited treatment options, and poor prognosis. Emerging evidence highlights estrogen receptor beta 1 (ERβ1) as a potential tumor suppressor in TNBC, influencing key oncogenic pathways such as cell proliferation, survival, angiogenesis, and apoptosis. In this regard, calcitriol (active vitamin D) and 17β-estradiol have been identified as key regulators of tumor behavior. Calcitriol shows strong anti-proliferative and pro-apoptotic effects, while the ability of 17β-estradiol to modulate tumor progression through ERβ1 signaling is context-dependent. This study aims to investigate the individual and combined effects of calcitriol and 17β-estradiol in ERβ1-expressing MDA-MB-468 TNBC cells, with a focus on their role in regulation of tumor progression, angiogenesis, and apoptosis. The findings provide novel insights into the potential therapeutic utility of targeting ERβ1 in TNBC. Methodology The MDA-MB-468 TNBC cells were treated with calcitriol (1-5 µM) and/or 17β-estradiol (100-500 nM). The effect on cell viability was assayed with the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) method. At the same time, immunoblot analysis investigated the time-dependent manner of ERβ1, epidermal growth factor receptor (EGFR), vascular epithelial growth factor (VEGF), and caspase-3. Results Both calcitriol and 17β-estradiol substantially decreased TNBC cell viability, with the highest level of cytotoxicity observed at 24 and 32 hours, respectively. The combination increased the amount of cell death. Immunoblots revealed lasting downregulation of ERβ1, EGFR, VEGF, and caspase-3 after calcitriol treatment. In comparison, 17β-estradiol demonstrated biphasic regulatory behavior for ERβ1, where ERβ1 was first downregulated, then partially recovered. The combination therapies produced more significant ERβ1 downregulation and heightened suppression of EGFR and VEGF, further enhancing their effects on TNBC progression. Conclusions This study aims to investigate the individual and combined effects of calcitriol and 17β-estradiol in ERβ1-expressing MDA-MB-468 TNBC cells, with a focus on their role in the regulation of tumor progression, angiogenesis, and apoptosis.
Collapse
Affiliation(s)
- Shankaramurthy K N
- Biochemistry, Bijapur Lingayat District Educational (BLDE) Shri B.M. Patil Medical College Hospital and Research Centre, Vijayapura, IND
| | - Praveenkumar Shetty
- Biochemistry, K S Hegde Medical Academy, NITTE (Deemed to be University), Mangaluru, IND
| | - Basavaraj Devaranagadi
- Biochemistry, Bijapur Lingayat District Educational (BLDE) Shri B.M. Patil Medical College Hospital and Research Centre, Vijayapura, IND
| | - Indira A Hundekari
- Biochemistry, Bijapur Lingayat District Educational (BLDE) Shri B.M. Patil Medical College Hospital and Research Centre, Vijayapura, IND
| |
Collapse
|
262
|
Fu D, Miao H, Wang Z, Yang C. Gynecomastia and its potential progression to male breast cancer: Mechanisms, genetic factors, and hormonal interactions. Crit Rev Oncol Hematol 2025; 208:104651. [PMID: 39909181 DOI: 10.1016/j.critrevonc.2025.104651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 01/20/2025] [Accepted: 01/31/2025] [Indexed: 02/07/2025] Open
Abstract
Gynecomastia is the most common breast condition in men, while male breast cancer remains relatively rare. This review explores the potential relationship between gynecomastia and male breast cancer, with a focus on the roles of hormonal imbalances, genetic factors, and molecular mechanisms in the progression of these conditions. While it remains controversial whether gynecomastia is a precancerous lesion for male breast cancer, this review summarizes the roles of estrogen and androgen receptors, the regulation of aromatase expression, and mutations in key genes such as BRCA1/2. These insights point to possible pathways by which gynecomastia could transition into male breast cancer. Additionally, hormones such as prolactin, insulin-like growth factor-1, and leptin may play significant roles in this progression. We provide an overview of the current understanding and identify key areas for further research, emphasizing the need for large-scale prospective studies to determine the causal relationship between gynecomastia and male breast cancer.
Collapse
Affiliation(s)
- Dingyi Fu
- Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China; Nanjing Medical University, Nanjing 211166, China
| | - Haoquan Miao
- Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China; Nanjing Medical University, Nanjing 211166, China
| | - Zhonglin Wang
- Department of General Surgery, The Second People's Hospital of Lianyungang, Lianyungang 222006, China
| | - Chuang Yang
- Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
263
|
Zhao L, Wu X, Li Q, Shen Y, Zeng S, Wang J, Liu Q. Inhibition of CCN5 Protects Against Apoptosis and Endoplasmic Reticulum Stress in Bisphenol A-Induced Sertoli Cells via p38/JNK MAPK Signaling Pathway. DNA Cell Biol 2025; 44:174-185. [PMID: 40184555 DOI: 10.1089/dna.2024.0247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2025] Open
Abstract
Bisphenol A (BPA) is the most common endocrine disruptor that has toxicity to the reproductive system and male infertility. However, the underlying mechanisms of BPA's toxicity to Sertoli cells remain poorly understood. Cellular communication network factor 5 (CCN5) is reported to regulate cell proliferation, apoptosis, and differentiation. Our study demonstrated a significant elevation of CCN5 expression in the testis of nonobstructive azoospermia patients and TM4 Sertoli cells exposed to BPA. Knockdown of CCN5 reduced apoptotic cells after BPA treatment, as determined by flow cytometry and terminal deoxynucleotidyl transferase dUTP nick end labeling assays. Cells exposed to BPA showed increased expressions of Bax and cleaved poly(ADP-ribose) polymerase, decreased expression of Bcl-2, as well as elevated activities of caspase-3 and caspase-9 in BPA-induced TM4 cells, which were reversed by CCN5 inhibition. Loss of CCN5 declined phosphorylation of protein kinase R-like endoplasmic reticulum kinase and eukaryotic translation initiation factor 2A and decreased activating transcription factor 4 and C/EBP-homologous protein in BPA-treated cells. Furthermore, silencing CCN5 blocked BPA-induced phosphorylation of p38 and c-Jun N-terminal kinase (JNK). Administration of anisomycin, a mitogen-activated protein kinase (MAPK) activator, reversed the effects of CCN5 knockdown on BPA-induced endoplasmic reticulum (ER) stress and apoptosis. Taken together, CCN5 promotes apoptosis and ER stress in Sertoli cells exposed to BPA by activating the p38/JNK MAPK signaling pathway.
Collapse
Affiliation(s)
- Lijiang Zhao
- Tianjin Medical University, Tianjin, China
- Department of Urology, Tianjin First Central Hospital, Tianjin, China
- Reproductive Medicine Center, Shanxi Children's Hospital, Shanxi Women and Children Hospital, Taiyuan, China
| | - Xueqing Wu
- Reproductive Medicine Center, Shanxi Children's Hospital, Shanxi Women and Children Hospital, Taiyuan, China
| | - Qiang Li
- Reproductive Medicine Center, Shanxi Children's Hospital, Shanxi Women and Children Hospital, Taiyuan, China
| | - Yan Shen
- Reproductive Medicine Center, Shanxi Children's Hospital, Shanxi Women and Children Hospital, Taiyuan, China
| | - Sheng Zeng
- Department of Urology, Tianjin First Central Hospital, Tianjin, China
| | - Jinbao Wang
- Reproductive Medicine Center, Shanxi Children's Hospital, Shanxi Women and Children Hospital, Taiyuan, China
| | - Qian Liu
- Department of Urology, Tianjin First Central Hospital, Tianjin, China
| |
Collapse
|
264
|
EFSA Panel on Additives and Products or Substances used in Animal Feed (FEEDAP), Villa RE, Azimonti G, Bonos E, Christensen H, Durjava M, Dusemund B, Gehring R, Glandorf B, Kouba M, López‐Alonso M, Marcon F, Nebbia C, Pechová A, Prieto‐Maradona M, Röhe I, Theodoridou K, Anguita M, Cionci NB, Innocenti ML, Kolona M, Tarrés‐Call J. Safety and efficacy of a feed additive consisting of l-lysine monohydrochloride produced with Corynebacterium glutamicumCGMCC 7.453 for all animal species (Eppen Europa SAS). EFSA J 2025; 23:e9345. [PMID: 40182014 PMCID: PMC11966234 DOI: 10.2903/j.efsa.2025.9345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025] Open
Abstract
Following a request from the European Commission, EFSA was asked to deliver a scientific opinion on the safety and efficacy of l-lysine monohydrochloride produced by fermentation with a genetically modified strain of Corynebacterium glutamicum (CGMCC 7.453) as nutritional feed additive for all animal species. Neither viable cells nor recombinant DNA of the production strain were detected in the final product. Therefore, the FEEDAP Panel concluded that the additive does not pose any safety concern regarding the production strain. The FEEDAP Panel concluded that the use of l-lysine HCl produced by fermentation with the strain C. glutamicum CGMCC 7.453 is safe for the target species when administered via feed. However, the FEEDAP Panel expressed concerns on the use of l-lysine HCl in water for drinking. The Panel concluded that the use of l-lysine HCl produced by fermentation with C. glutamicum CGMCC 7.453 in animal nutrition is considered safe for the consumers and for the environment. With regards user safety, the additive should be considered irritant to skin, eyes and the respiratory tract. Any exposure to the additive is a risk. l-Lysine HCl is considered as efficacious source of the essential amino acid l-lysine for non-ruminant animal species. For the supplemental l-lysine to be as efficacious in ruminants as in non-ruminant species, it would require protection against degradation in the rumen.
Collapse
|
265
|
EFSA Panel on Additives and Products or Substances used in Animal Feed (FEEDAP), Villa RE, Azimonti G, Bonos E, Christensen H, Durjava M, Dusemund B, Gehring R, Glandorf B, Kouba M, López‐Alonso M, Marcon F, Nebbia C, Pechová A, Prieto‐Maradona M, Röhe I, Theodoridou K, Galobart J, Holczknecht O, Pizzo F, Radovnikovic A, Vettori MV, Amaduzzi A. Safety and efficacy of the feed additive consisting of clinoptilolite of sedimentary origin for all animal species for the renewal of its authorisation (ZEOCEM, a.s.). EFSA J 2025; 23:e9364. [PMID: 40226503 PMCID: PMC11986687 DOI: 10.2903/j.efsa.2025.9364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2025] Open
Abstract
Following a request from the European Commission, the Panel on Additives and Products or Substances used in Animal Feed (FEEDAP) was asked to deliver a scientific opinion on the assessment of the application for renewal of authorisation of clinoptilolite of sedimentary origin as a technological feed additive (binder and anticaking agents) for all animal species. The applicant provided evidence that the additive currently on the market complies with the existing conditions of authorisation. The Panel concludes that clinoptilolite of sedimentary origin remains safe for all animal species, consumers and the environment under the authorised conditions of use. Regarding user safety, the additive is not a skin nor an eye irritant but should be considered a skin and respiratory sensitiser. Inhalation and dermal exposure are considered a risk. There is no need for assessing the efficacy of the additive in the context of the renewal of the authorisation.
Collapse
|
266
|
Syngkli S, Singh SK, Rani RM, Das B. Functional and Biochemical Analyses of Glycerol Kinase and Glycerol 3-phosphate Dehydrogenase in HEK293 Cells. Protein J 2025; 44:231-244. [PMID: 39987391 DOI: 10.1007/s10930-025-10252-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2025] [Indexed: 02/24/2025]
Abstract
Type 2 diabetes mellitus (T2DM) is a chronic metabolic disorder and its concurrent presence with chronic kidney disease (CKD) is a significant concern. Glycerol kinase (GK) and glycerol 3-phosphate shuttle enzymes (cGPDH and mGPDH) facilitate the regulation of endogenous glucose production in many cell lines. This research investigates the functions of GK, cGPDH, and mGPDH in HEK293 cells. Standard protocols were employed to assess enzyme activity, mRNA- and protein-expression, glucose uptake, and production. Homology modeling and molecular docking were employed to elucidate interactions of genistein and metformin with these enzymes. The secondary structures of GK, cGPDH and mGPDH and the thermal stability of cGPDH and mGPDH were analyzed by CD spectra. Genistein inhibited GK activity by 40%, while metformin decreased cGPDH and mGPDH activity by 58% and 55%, respectively, in HEK293 cells. Nonetheless, the expression levels of mRNA and protein remained unaltered. Genistein and metformin inhibited HEK293 glucose production by 0.46-fold and 0.63-fold, respectively. Genistein reduced glucose uptake by 0.26-fold, while metformin increased it by 0.51-fold. Genistein allosterically interacted with GK with a CDocker energy of -27.71, while metformin interacted with Gln295 and Lys296 of the catalytic loop of cGPDH and the FAD+ binding domain of mGPDH, yielding CDocker energies of -11.12 and -13.34, respectively. This study indicated the role of genistein and metformin on GK, cGPDH, and mGPDH in HEK293 cells.
Collapse
Affiliation(s)
- Superior Syngkli
- Department of Zoology, North-Eastern Hill University, Shillong, 793022, India
| | - Sumit K Singh
- Department of Zoology, North-Eastern Hill University, Shillong, 793022, India
| | - Riva M Rani
- Department of Zoology, North-Eastern Hill University, Shillong, 793022, India
| | - Bidyadhar Das
- Department of Zoology, North-Eastern Hill University, Shillong, 793022, India.
| |
Collapse
|
267
|
Wang Y, Li N, Guan W, Wang D. Controversy and multiple roles of the solitary nucleus receptor Nur77 in disease and physiology. FASEB J 2025; 39:e70468. [PMID: 40079203 PMCID: PMC11904867 DOI: 10.1096/fj.202402775rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 02/27/2025] [Accepted: 03/07/2025] [Indexed: 03/14/2025]
Abstract
Neuron-derived clone 77 (Nur77), a member of the orphan nuclear receptor family, is expressed and activated rapidly in response to diverse physiological and pathological stimuli. It exerts complex biological functions, including roles in the nervous system, genome integrity, cell differentiation, homeostasis, oxidative stress, autophagy, aging, and infection. Recent studies suggest that Nur77 agonists alleviate symptoms of neurodegenerative diseases, highlighting its potential as a therapeutic target in such conditions. In cancer, Nur77 demonstrates dual roles, acting as both a tumor suppressor and promoter, depending on the cancer type and stage, making it a controversial yet promising anticancer target. This review provides a structured analysis of the functions of Nur77, focusing on its physiological and pathological roles, therapeutic potential, and existing controversies. Emphasis is placed on its emerging applications in neurodegenerative diseases and cancer, offering key insights for future research and clinical translation.
Collapse
Affiliation(s)
- Yanteng Wang
- Department of Gerontology and GeriatricsShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Na Li
- Department of Gerontology and GeriatricsShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Wenwei Guan
- Department of Gerontology and GeriatricsShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Difei Wang
- Department of Gerontology and GeriatricsShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| |
Collapse
|
268
|
Gao X, Liu G, Zhao Z, Tang Y, Hui H, Wang C, Li D, Ma Y, Sun Z, Zhou Y. Arsenic enhances cervical cancer cell radiosensitivity by suppressing the DNA damage repair pathway. Transl Cancer Res 2025; 14:2078-2094. [PMID: 40225006 PMCID: PMC11985179 DOI: 10.21037/tcr-2025-450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Accepted: 03/19/2025] [Indexed: 04/15/2025]
Abstract
Background Concurrent chemoradiotherapy (CCRT) is a primary treatment for cervical cancer (CC) and combines chemotherapy and radiation therapy to target cancer cells effectively. However, despite its benefits, it also involves a high risk of recurrence and metastasis, partly due to the resistance of some cancer cells to the treatment. Additionally, CCRT can cause various treatment-related adverse reactions, such as gastrointestinal issues, bone marrow suppression, and skin reactions, which can negatively impact patients' quality of life. Therefore, there is a compelling need to develop more effective treatment strategies that can improve the outcomes of CCRT while minimizing its side effects. This study aimed to investigate the radiosensitizing effects of arsenic trioxide (ATO) on CC and explore its underlying molecular mechanisms. Methods We conducted both in vitro and in vivo experiments to evaluate the radio-sensitizing properties of ATO. The in vitro effects of ATO were assessed using clonogenic assay, while in vivo effects were evaluated using a xenograft model. Then cell viability, cell cycle, and apoptosis were assessed by Cell Counting Kit-8 (CCK-8) assay and flow cytometry. RNA sequencing was performed to identify the differentially expressed genes. Finally, mRNA and protein expressions of key hub genes were analyzed by quantitative real-time polymerase chain reaction (qRT-PCR) and western blot. Western blot, immunofluorescence, and RNA sequencing analyzed molecular mechanisms. Results ATO significantly enhanced the radiosensitivity of CC cells, as evidenced by reduced colony formation in vitro and inhibited tumor growth in vivo. This enhancement was achieved by impairing the DNA damage repair pathway, specifically through the downregulation of key proteins such as breast cancer 1 (BRCA1) and bloom syndrome protein (BLM). Notably, overexpression of BRCA1 or BLM substantially mitigated ATO's radiosensitizing effects. Conclusions This study demonstrates that ATO exhibits radiosensitizing effects on CC by inhibiting DNA damage repair. These findings provide theoretical and experimental support for using ATO as a radiosensitizer in CC therapy, potentially leading to improved treatment outcomes, reduced recurrence rates, and enhanced patient survival. Future research should focus on optimizing ATO's dosage and timing as well as evaluating its long-term safety and efficacy in clinical settings.
Collapse
Affiliation(s)
- Xingxing Gao
- The Affiliated Xuzhou Clinical College of Xuzhou Medical University, Xuzhou, China
| | - Genyun Liu
- The Affiliated Xuzhou Clinical College of Xuzhou Medical University, Xuzhou, China
| | - Zimu Zhao
- Department of Pathology, School of Basic Medical Sciences, Xuzhou Key Laboratory of Clinical and Experimental Pathology, Xuzhou Medical University, Xuzhou, China
| | - Yi Tang
- The Affiliated Xuzhou Clinical College of Xuzhou Medical University, Xuzhou, China
| | - Hui Hui
- The Affiliated Xuzhou Clinical College of Xuzhou Medical University, Xuzhou, China
| | - Chaoqun Wang
- Department of Pathology, School of Basic Medical Sciences, Xuzhou Key Laboratory of Clinical and Experimental Pathology, Xuzhou Medical University, Xuzhou, China
| | - Danhua Li
- Department of Pathology, School of Basic Medical Sciences, Xuzhou Key Laboratory of Clinical and Experimental Pathology, Xuzhou Medical University, Xuzhou, China
| | - Yu Ma
- Department of Pathology, School of Basic Medical Sciences, Xuzhou Key Laboratory of Clinical and Experimental Pathology, Xuzhou Medical University, Xuzhou, China
| | - Zhuo Sun
- Department of Pathology, School of Basic Medical Sciences, Xuzhou Key Laboratory of Clinical and Experimental Pathology, Xuzhou Medical University, Xuzhou, China
| | - Yun Zhou
- The Affiliated Xuzhou Clinical College of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
269
|
Sanfilippo C, Castrogiovanni P, Imbesi R, Vecchio M, Vinciguerra M, Blennow K, Zetterberg H, Di Rosa M. Sex-specific modulation of FOLR1 and its cycle enzyme genes in Alzheimer's disease brain regions. Metab Brain Dis 2025; 40:163. [PMID: 40153031 DOI: 10.1007/s11011-025-01578-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 03/13/2025] [Indexed: 03/30/2025]
Abstract
Alzheimer's disease (AD) is the most common form of dementia, characterized by progressive cognitive and functional decline. Its incidence increases significantly with age and is more prevalent in women than men. We investigated the folate receptor alpha (FOLR1) gene expression levels in the central nervous system (CNS) of AD and non-demented healthy control (NDHC) subjects. Our cohort included 3,946 samples: 2,391 NDHC and 1,555 AD patients, stratified by brain region, age, and sex. Interestingly, a significant increase in FOLR1 expression was observed only in females with AD compared to NDHC females. Furthermore, we found that FOLR1 expression was differentially increased in the prefrontal cortex (PFC) and diencephalon (DIE) only in AD females. Moreover, in females, genes involved in the folic acid (FA) cycle that drives DNA synthesis were significantly modulated. In contrast, in males, downregulation of TYMS effectively blocks the completion of the cycle, thereby preventing downstream DNA synthesis. Tissue Transcriptome Deconvolution (TTD) analysis revealed astrocytes and endothelial cells associated with FOLR1 expression in both AD males and females. Gene Ontology analysis supported these findings, showing enrichment in processes aligned with these cell types. Positive correlations between brain FOLR1 expression and markers for astrocytes (glial fibrillary acidic protein) and endothelial cells (CD31) provided further validation. Our findings suggest a potential role for sex-dependent FOLR1 expression and its association with specific brain regions and cellular processes in AD.
Collapse
Affiliation(s)
- Cristina Sanfilippo
- Neurologic Unit, AOU "Policlinico-San Marco", Department of Medical, Surgical Sciences and Advanced Technologies, GF, Ingrassia, University of Catania, Via Santa Sofia n.78, Catania, Sicily, 95100, Italy
| | - Paola Castrogiovanni
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Catania, Italy
| | - Rosa Imbesi
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Catania, Italy
| | - Michele Vecchio
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Manlio Vinciguerra
- Department of Translational Stem Cell Biology, Research Institute, Medical University Varna, Varna, Bulgaria
- Liverpool Centre for Cardiovascular Science, Faculty of Health, Liverpool John Moores University, Liverpool, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- UK Dementia Research Institute at UCL, London, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer'S Disease Research Center, School of Medicine and Public Health, University of Wisconsin, University of Wisconsin-Madison, Madison, WI, USA
| | - Michelino Di Rosa
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Catania, Italy.
| |
Collapse
|
270
|
Khandan M, Khazeei Tabari MA, Rahimi SM, Hassani M, Bagheri A. The effects of flavonoid baicalein on miRNA expressions in cancer: a systematic review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04078-y. [PMID: 40153015 DOI: 10.1007/s00210-025-04078-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 03/19/2025] [Indexed: 03/30/2025]
Abstract
Baicalein from Scutellaria baicalensis influences miRNA expression in various cancers, affecting key signaling pathways (PI3K/AKT, Wnt/β-catenin, mTOR) and processes like tumor growth, apoptosis, and metastasis. miRNAs, as small non-coding RNAs, play crucial roles in the cancer pathogenesis-associated gene regulations. This study is aimed at systematically reviewing the effects of baicalein on miRNA expression in various cancers. A comprehensive systematic review was conducted following PRISMA guidelines to investigate the impact of baicalein on miRNA expression in cancer. Databases including PubMed, Scopus, and Web of Science were systematically searched using key search terms. Inclusion criteria encompassed studies reporting changes in miRNA expression following baicalein treatment in cancer cell lines and animal models. Data extraction and risk of bias assessment based on SYRCLE's risk of bias tool were performed to ensure methodological rigor and reliability of the findings. Fifteen studies meeting the inclusion criteria were included in the systematic review. Baicalein impacts miRNA expression in cancers like hepatocellular carcinoma, breast, cervical, ovarian, and gastric cancers, suggesting its potential as a multi-cancer therapeutic. Baicalein regulates tumor-related genes (HDAC10, MDM2, Bcl-2/Bax, and Cyclin E1) and signaling molecules (AKT, FOXO3α), affecting cell viability, apoptosis, and cell cycle, indicating targeted therapeutic potential. In vitro and in vivo studies show baicalein inhibits tumor growth, enhances apoptosis, and regulates cell proliferation, supporting its anticancer effects. Baicalein exhibits potential in modulating miRNA expression in cancer, offering avenues for therapeutic intervention. However, methodological rigor in future studies is essential to enhance the reliability and validity of findings. Comprehensive understanding of baicalein's effects on miRNA expression holds promise for developing novel cancer treatment strategies.
Collapse
Affiliation(s)
| | | | | | - Mahmoud Hassani
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abouzar Bagheri
- Imam Khomeini Hospital, Mazandaran University of Medical Sciences, Sari, Iran.
- Immunogenetics Research Center, Department of Clinical Biochemistry and Medical Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
271
|
Gong P, Zhao L, Ma Y, Shu Q, Sun H, Lu J, Meng F, Wan F. AHR Agonist ITE Boosted PD1 Antibody's Effects by Inhibiting Myeloid-Derived Cells Suppressive Cells in an Orthotopic Mouse Glioma Model. Pharmaceuticals (Basel) 2025; 18:471. [PMID: 40283908 PMCID: PMC12030425 DOI: 10.3390/ph18040471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 12/25/2024] [Accepted: 01/27/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Glioblastoma is "cold". Consequently, immune checkpoint blockade therapy has failed to improve patients' survival, which is negatively correlated with patients' peripheral MDSC counts. AHR is known to mediate immune-suppressive functions of certain tryptophan metabolites such as kynurenine; yet, there lack of reports on how AHR agonists affect glioma immunity. Methods/Objectives: We hypothesized that ITE could synergize with PD1 antibody as AHR is one major node of immune-suppressive pathways, and tested it using an immune-competent mouse glioma model. Results: The combination of ITE+PD1 antibody glioma MDSC was significantly reduced, along with increased infiltration of the CD4-CD8+ and CD4+CD8+ T cells, leading to extended mouse survival. ITE treatment alone significantly reduces the infiltration of CD11b+Ly6G+Ly6Clo cells, namely PMN-MDSCs, and neutrophils, while the combination with PD1 antibody significantly reduces all MDSCs plus neutrophils. The presence of ITE inhibits the expression of IL11 and the in vitro induction of MDSCs from mouse PBMCs by IL11. The identification of the ITE-AHR-IL11-MDSC pathway provides more mechanistic insights into AHR's effects. The fact that ITE, which is otherwise immune-suppressive, can activate immunity in glioma indicates that searching for drugs targeting AHR should go beyond antagonists.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Fanhua Meng
- Tumor Molecular Pharmacology Laboratory, College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010011, China; (P.G.); (L.Z.); (Y.M.); (Q.S.); (H.S.); (J.L.)
| | - Fang Wan
- Tumor Molecular Pharmacology Laboratory, College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010011, China; (P.G.); (L.Z.); (Y.M.); (Q.S.); (H.S.); (J.L.)
| |
Collapse
|
272
|
Patnam S, Singh AD, Ali MS, Thakur BK, Rengan AK, Manda SV. Development and In Vitro Characterization of Milk-Derived Extracellular Vesicle-Mithramycin Formulations for Potential Glioma Therapy. Mol Pharm 2025. [PMID: 40138182 DOI: 10.1021/acs.molpharmaceut.4c01189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Glioblastoma (GBM) is a highly aggressive brain tumor with resistance to conventional therapies. Mithramycin (Mit-A), a potent antitumor agent, has shown promise in several tumor types including, GBM. However, its clinical application is limited by toxicity. To address this, we explored the use of milk-derived extracellular vesicles (mEVs) as a delivery system to enhance the therapeutic efficacy of Mit-A. In this study, mEVs were isolated using a 3000 PEG precipitation method and confirmed their size, morphology, and stability through dynamic light scattering (DLS), transmission electron microscopy (TEM), and atomic force microscopy (AFM). The isolated vesicles with a size of 125.6 ± 2.78 nm, a polydispersity index (PDI) of 0.083 ± 0.02, and a ζ-potential of 15 ± 0.57 mV. The presence of typical EV markers such as TSG101, HSP70, and CD63 confirmed their purity. Encapsulation of Mit-A within mEVs led to a slight increase in size to 131.8 ± 6.9 nm, a PDI of 0.081 ± 0.006, and a decrease in ζ-potential to -17 ± 2.0 mV, with an encapsulation efficiency of 58% by the freeze-thaw method. The in vitro transepithelial transport assay revealed that mEV(Mit-A) transported Mit-A more effectively than free Mit-A. The mEV(Mit-A) formulation demonstrated excellent stability in simulated salivary and gastrointestinal fluids, with a sustained release of Mit-A observed over 24 h in vitro in PBS (pH 6.8). Furthermore, mEV(Mit-A) formulations significantly inhibited glioma cell growth, and migration, and induced apoptosis, showing a 2-fold lower IC50 than free Mit-A, indicating superior efficacy. These findings suggest that mEVs represent a promising delivery vehicle for Mit-A, enhancing its potential as an effective treatment for glioblastoma.
Collapse
Affiliation(s)
- Sreekanth Patnam
- Apollo Hospitals Educational and Research Foundation (AHERF), Hyderabad, Telangana 500096, India
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Telangana 502284, India
- Exomed Therapeutics Private Ltd., Hyderabad, Telangana 500096, India
| | - Anula Divyash Singh
- Apollo Hospitals Educational and Research Foundation (AHERF), Hyderabad, Telangana 500096, India
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Telangana 502284, India
| | - Mohammad Sadik Ali
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Telangana 502284, India
| | - Basant Kumar Thakur
- Department of Pediatrics III, University Hospital Essen, Hufelandstr. 55, 45122 Essen, Germany
| | - Aravind Kumar Rengan
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Telangana 502284, India
| | - Sasidhar Venkata Manda
- Apollo Hospitals Educational and Research Foundation (AHERF), Hyderabad, Telangana 500096, India
- UrvogelBio Private Ltd., Hyderabad, Telangana 500096, India
| |
Collapse
|
273
|
Shao B, Wang HD, Ren SH, Chen Q, Wang ZB, Xu YN, Liu T, Sun CL, Xiao YY, Jiang HY, Li YC, Zhao PY, Yang GM, Liu X, Ren YF, Wang H. Exosomes derived from a mesenchymal-like endometrial regenerative cells ameliorate renal ischemia reperfusion injury through delivery of CD73. Stem Cell Res Ther 2025; 16:148. [PMID: 40140882 PMCID: PMC11948919 DOI: 10.1186/s13287-025-04275-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Renal ischemia reperfusion (I/R) injury is a major contributor to graft dysfunction and inflammation leading to graft loss. The deregulation of purinergic signaling has been implicated in the pathogenesis of renal I/R injury. CD73 and the generation of adenosine during purine metabolism to protect against renal I/R injury. A mesenchymal-like endometrial regenerative cell (ERC) has demonstrated a significant therapeutic effect on renal I/R injury. CD73 is a phenotypic marker of human endometrial regenerative cell exosomes (ERC-Exo). However, its immunosuppressive function in regulating purinergic metabolism has been largely neglected. Here, we investigate the protective effects and mechanism of ERC-Exo against renal I/R injury. METHODS Lentivirus-mediated CRISPR-Cas9 technology was employed to obtain CD73-specific knockout ERC-Exo (CD73-/-ERC-Exo). C57BL/6 mice who underwent unilateral ureteral obstruction were divided into the Untreated, ERC-Exo-treated, and CD73-/-ERC-Exo-treated groups. Renal function and pathological injury were assessed 3 days after renal reperfusion. The infiltration of CD4+ T cells and macrophages was analyzed by flow cytometry and immunofluorescence staining in kidneys. CD73-mediated immunosuppressive activity of ERC-Exo was investigated by bone marrow-derived macrophages (BMDM) co-culture assay in vitro. Flow cytometry determined macrophage polarization. ELISA and Treg proliferation assays detected the function of macrophages. Furthermore, the role of the MAPK pathway in CD73-positive Exo-induced macrophage polarization was also elucidated. RESULTS Compared with Untreated and CD73-/-ERC-Exo-treated groups, CD73-positive Exo effectively improved the serum creatinine (sCr), blood urea nitrogen (BUN), and necrosis and detachment of tubular epithelial cells, necrosis and proteinaceous casts induced by ischemia. CD73 improved the capacity of ERC-Exo on CD4+ T cell differentiation in the renal immune microenvironment. Surprisingly, ERC-Exosomal CD73 significantly decreased the populations of M1 cells but increased the proportions of M2 in kidneys. Furthermore, CD73-positive Exo markedly reduced the levels of proinflammatory cytokines (IL-1β, IL-6, and TNF-α) and increased anti-inflammatory factors (IL-10) level in kidneys. ERC-Exosomal CD73 improved macrophage immunoregulatory function associated with the MAPK pathway (including ERK1/2 and p38 pathways), which exerted a potent therapeutic effect against renal I/R. CONCLUSIONS These data collected insight into how ERC-Exo facilitated the hydrolysis of proinflammatory ATP to immunosuppressive ADO via CD73. CD73 is a critical modulator of the MAPK signaling pathway, inducing a polarization shift of macrophages towards an anti-inflammatory phenotype. This study highlights the significance of ERC-Exosomal CD73 in contributing to the therapeutic effects against renal I/R.
Collapse
Affiliation(s)
- Bo Shao
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Hong-da Wang
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Shao-Hua Ren
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
- Department of General Surgery, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Qiang Chen
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhao-Bo Wang
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Yi-Ni Xu
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Tong Liu
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Cheng-Lu Sun
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Yi-Yi Xiao
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Hong-Yu Jiang
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Yi-Cheng Li
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Peng-Yu Zhao
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Guang-Mei Yang
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Xu Liu
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Yu-Fan Ren
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Hao Wang
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China.
- Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin, China.
| |
Collapse
|
274
|
Pérez Escriva P, Correia Tavares Bernardino C, Letellier E. De-coding the complex role of microbial metabolites in cancer. Cell Rep 2025; 44:115358. [PMID: 40023841 DOI: 10.1016/j.celrep.2025.115358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 12/11/2024] [Accepted: 02/06/2025] [Indexed: 03/04/2025] Open
Abstract
The human microbiome, an intricate ecosystem of trillions of microbes residing across various body sites, significantly influences cancer, a leading cause of morbidity and mortality worldwide. Recent studies have illuminated the microbiome's pivotal role in cancer development, either through direct cellular interactions or by secreting bioactive compounds such as metabolites. Microbial metabolites contribute to cancer initiation through mechanisms such as DNA damage, epithelial barrier dysfunction, and chronic inflammation. Furthermore, microbial metabolites exert dual roles on cancer progression and response to therapy by modulating cellular metabolism, gene expression, and signaling pathways. Understanding these complex interactions is vital for devising new therapeutic strategies. This review highlights microbial metabolites as promising targets for cancer prevention and treatment, emphasizing their impact on therapy responses and underscoring the need for further research into their roles in metastasis and therapy resistance.
Collapse
Affiliation(s)
- Pau Pérez Escriva
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Catarina Correia Tavares Bernardino
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Elisabeth Letellier
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.
| |
Collapse
|
275
|
Guo H, Liu C, Wu K, Li Y, Zhang Z, Chen F. Single-cell RNA sequencing reveals an IL1R2+Treg subset driving immunosuppressive microenvironment in HNSCC. Cancer Immunol Immunother 2025; 74:159. [PMID: 40131478 PMCID: PMC11936857 DOI: 10.1007/s00262-025-04015-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 03/05/2025] [Indexed: 03/27/2025]
Abstract
Regulatory T cells (Tregs) play an immunosuppressive role in tumor microenvironment (TME) in various of cancer types. However, how different Treg subsets influence and effect on head and neck squamous cell carcinoma (HNSCC) remain unclear. Here, using single-cell RNA sequencing (scRNA-seq), we identified an IL1R2+Treg subset which promoted the progression of HNSCC. Via tissue microassay (TMA) and enzyme-linked immunosorbent assay (ELISA), we verified the clinical diagnostic value of the IL1R2+Treg and soluble IL1R2 (sIL1R2). In addition, we constructed tumor-bearing mouse models to explore the antitumor effects of combined targeting IL1R2 and CTLA4. For mechanism, we found IL-1β promoted the expression of IL1R2 and CTLA4 in Tregs, and upregulated CTLA4 though NR4A1 translocation. These results revealed that IL1R2+Treg and serum IL1R2 level had potential diagnostic and prognostic value of HNSCC and combined targeting of IL1R2 and CTLA4 might be an effective strategy to inhibit tumor progression.
Collapse
Affiliation(s)
- Haiyan Guo
- Department of Clinical Immunology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Chun Liu
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kun Wu
- Department of Oral and Maxillofacial Surgery, Second Xiangya Hospital of Central South University, Changsha, China
| | - Yan Li
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhen Zhang
- Department of Clinical Immunology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, China.
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Diagnosis and Treatment Innovation Center for Cancer, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Fuxiang Chen
- Department of Clinical Immunology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, China.
- Faculty of Medical Laboratory Science, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
276
|
Hamilton AM, Srivastava VK, Hiney JK, Dees WL, Dearth RK. Manganese-induced Precocious Puberty Alters Mammary Epithelial Cell Proliferation in Female Rats. Endocrinology 2025; 166:bqaf052. [PMID: 40105700 PMCID: PMC12006721 DOI: 10.1210/endocr/bqaf052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 01/02/2025] [Accepted: 03/17/2025] [Indexed: 03/20/2025]
Abstract
Precocious puberty (PP) is an established breast cancer risk factor. In the normal mammary gland, hormone receptor-positive (HR+) cells rarely proliferate. In breast cancer, proliferating epithelial cells are often HR+. It is not known if PP can modify this population of proliferating HR+ cells. Previously, we established a manganese-induced precocious puberty (MnPP) model to study the effects of PP on mammary gland development in female rats. Here, we characterized the distribution of HR+ proliferating mammary epithelial cells in prepubertal and adult rodents, in association with precocious puberty. Female rats were exposed daily to 10 mg/kg manganese chloride or saline (control) from postnatal day (PND) 12 to PND 30. Mammary glands were collected on PNDs 30 and 120, processed for western blot analysis and double immunofluorescence staining for proliferating cell nuclear antigen and progesterone receptor or estrogen receptor. MnPP increased the percentage of HR+ mammary epithelial cells coexpressing proliferating cell nuclear antigen relative to normally developed controls at PND 30. This correlated with increased expression of estrogen receptor-regulated proteins in MnPP mammary glands relative to controls at PND 30, including FOXA1, AREG, and c-Myc. Conversely, at PND 120 relative to PND 30, proliferating HR+ cells remained chronically elevated in MnPP mammary glands at PND 120, which coincided with decreased expression of cell-cycle regulator, p27, and increased expression of progesterone receptor-regulated markers, EREG and sp1. Collectively, these results suggest early puberty alters steroidal regulation of classic proliferative mechanisms in the prepubertal gland with increased prevalence of high-risk proliferating HR+ cells.
Collapse
MESH Headings
- Animals
- Female
- Cell Proliferation/drug effects
- Epithelial Cells/drug effects
- Epithelial Cells/metabolism
- Mammary Glands, Animal/drug effects
- Mammary Glands, Animal/metabolism
- Mammary Glands, Animal/cytology
- Mammary Glands, Animal/growth & development
- Mammary Glands, Animal/pathology
- Rats
- Puberty, Precocious/chemically induced
- Puberty, Precocious/metabolism
- Puberty, Precocious/pathology
- Rats, Sprague-Dawley
- Receptors, Progesterone/metabolism
- Proliferating Cell Nuclear Antigen/metabolism
- Receptors, Estrogen/metabolism
- Sexual Maturation/drug effects
- Manganese
- Chlorides
- Manganese Compounds
Collapse
Affiliation(s)
- Alina M Hamilton
- School of Integrative Biological and Chemical Sciences, College of Sciences, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27606, USA (Current)
| | - Vinod K Srivastava
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843-4458, USA
| | - Jill K Hiney
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843-4458, USA
| | - William L Dees
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843-4458, USA
| | - Robert K Dearth
- School of Integrative Biological and Chemical Sciences, College of Sciences, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
| |
Collapse
|
277
|
Li Y, Izhar T, Kanekiyo T. HDAC3 as an Emerging Therapeutic Target for Alzheimer's Disease and other Neurological Disorders. Mol Neurobiol 2025:10.1007/s12035-025-04866-w. [PMID: 40126601 DOI: 10.1007/s12035-025-04866-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia in the aged population. Histone acetylation is a major epigenetic mechanism linked to memory formation and cognitive function. Histone deacetylases (HDACs) are responsible for the deacetylation of lysine residues in histone proteins. Although pan-HDAC inhibitors are effective in ameliorating AD phenotypes in preclinical models, they are associated with potential unfavorable adverse effects and barely translated into clinical trials. Therefore, the development of novel HDAC inhibitors with a well isoform-selectivity has been desired in AD drug discovery. Among various HDAC isoforms, HDAC3 is highly expressed in neurons and exhibits detrimental effects on synaptic plasticity and cognitive function. Moreover, HDAC3 provokes neuroinflammation and neurotoxicity and contributes to AD pathogenesis. In this review, we highlight HDAC3 as an attractive therapeutic target for disease-modifying therapy in AD. In addition, we discuss the therapeutic potential of HDAC3 inhibitors in other neurological disorders.
Collapse
Affiliation(s)
- Yonghe Li
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA.
| | - Taha Izhar
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Takahisa Kanekiyo
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| |
Collapse
|
278
|
Wang R, Wang Q, Liao J, Yu X, Li W. Piperlongumine overcomes osimertinib resistance via governing ubiquitination-modulated Sp1 turnover. JCI Insight 2025; 10:e186165. [PMID: 40125551 PMCID: PMC11949057 DOI: 10.1172/jci.insight.186165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 01/31/2025] [Indexed: 03/25/2025] Open
Abstract
Non-small cell lung cancer (NSCLC) is a common cause of cancer-related deaths worldwide, and its incidence has been increasing in recent years. While targeted therapies like osimertinib, an epidermal growth factor receptor tyrosine kinase inhibitor, have brought about notable improvements in patient outcomes for advanced NSCLC, the challenge of acquired drug resistance persists. Here, we found that cellular mesenchymal-epithelial transition factor (c-Met) was highly expressed in osimertinib-resistant cells, and depletion of c-Met markedly inhibited the growth of osimertinib-resistant cells ex vivo and in vivo, suggesting that c-Met is a potential target to address osimertinib resistance. Through a screening process using a natural product compound library, we identified piperlongumine as a potent inhibitor to overcome osimertinib resistance. Furthermore, the combined treatment of piperlongumine and osimertinib exhibited robust antitumor effects in resistant cells, partially restoring their sensitivity to osimertinib. Additionally, we discovered that piperlongumine could enhance the interaction between E3 ligase RNF4 and Sp1, inhibit the phosphorylation of Sp1 at Thr739, facilitate the ubiquitination and degradation of Sp1, lead to c-Met destabilization, and trigger intrinsic apoptosis in resistant cells. In summary, our study sheds light on the potential of piperlongumine in overcoming osimertinib resistance, offering new strategies and perspectives for the clinical management of drug-resistant NSCLC.
Collapse
Affiliation(s)
| | - Qiang Wang
- NHC Key Laboratory of Translational Research on Transplantation Medicine, Department of Transplant Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | | | - Xinfang Yu
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis of National Health Commission, Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha, China
| | - Wei Li
- Department of Radiology and
| |
Collapse
|
279
|
Li Q, Yang X, Li T. Natural flavonoids from herbs and nutraceuticals as ferroptosis inhibitors in central nervous system diseases: current preclinical evidence and future perspectives. Front Pharmacol 2025; 16:1570069. [PMID: 40196367 PMCID: PMC11973303 DOI: 10.3389/fphar.2025.1570069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Accepted: 02/24/2025] [Indexed: 04/09/2025] Open
Abstract
Flavonoids are a class of important polyphenolic compounds, renowned for their antioxidant properties. However, recent studies have uncovered an additional function of these natural flavonoids: their ability to inhibit ferroptosis. Ferroptosis is a key mechanism driving cell death in central nervous system (CNS) diseases, including both acute injuries and chronic neurodegenerative disorders, characterized by iron overload-induced lipid peroxidation and dysfunction of the antioxidant defense system. This review discusses the therapeutic potential of natural flavonoids from herbs and nutraceuticals as ferroptosis inhibitors in CNS diseases, focusing on their molecular mechanisms, summarizing findings from preclinical animal models, and providing insights for clinical translation. We specifically highlight natural flavonoids such as Baicalin, Baicalein, Chrysin, Vitexin, Galangin, Quercetin, Isoquercetin, Eriodictyol, Proanthocyanidin, (-)-epigallocatechin-3-gallate, Dihydromyricetin, Soybean Isoflavones, Calycosin, Icariside II, and Safflower Yellow, which have shown promising results in animal models of acute CNS injuries, including ischemic stroke, cerebral ischemia-reperfusion injury, intracerebral hemorrhage, subarachnoid hemorrhage, traumatic brain injury, and spinal cord injury. Among these, Baicalin and its precursor Baicalein stand out due to extensive research and favorable outcomes in acute injury models. Mechanistically, these flavonoids not only regulate the Nrf2/ARE pathway and activate GPX4/GSH-related antioxidant pathways but also modulate iron metabolism proteins, thereby alleviating iron overload and inhibiting ferroptosis. While flavonoids show promise as ferroptosis inhibitors for CNS diseases, especially in acute injury settings, further studies are needed to evaluate their efficacy, safety, pharmacokinetics, and blood-brain barrier penetration for clinical application.
Collapse
Affiliation(s)
- Qiuhe Li
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaohang Yang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Tiegang Li
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
280
|
Wang Y, Quan L, Zheng X, Hu Q, Huang X, Pu Y, Xie G, Peng Q. Indole compounds from fermented soybean products activate the aryl hydrocarbon receptor to reduce liver injury. NPJ Sci Food 2025; 9:38. [PMID: 40122901 PMCID: PMC11930980 DOI: 10.1038/s41538-025-00404-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 03/03/2025] [Indexed: 03/25/2025] Open
Abstract
The consumption of stinky tofu, a traditional fermented soybean product from China, elevates the concentrations of indole and trimethylindole in murine feces and increases the levels of indole in serum, as well as indole in the liver. These hepatic compounds act as ligands for the Aryl Hydrocarbon Receptor (AHR), triggering activation of this receptor, which subsequently enhances the expression of the enzyme cytochrome P450 (CYP) 1A1. This upregulation diminishes the levels of pro-inflammatory cytokines, thereby attenuating alcohol-induced liver injury. This study underscores the potential of dietary indole from stinky tofu to mitigate Alcoholic liver disease (ALD), laying a foundation for the development of functional foods and novel treatment strategies for ALD.
Collapse
Affiliation(s)
- Yanyun Wang
- College of Life Science, Leshan Normal University, Leshan, China
| | - Leping Quan
- National Engineering Research Center for Chinese CRW (branch center), School of Life and Environmental Sciences, Shaoxing University, Shaoxing, China
| | - Xiaomin Zheng
- Wuxi Maternity and Child Health Care Hospital, Affiliated Women's Hospital of Jiangnan University, Wuxi, China
| | - Qiang Hu
- College of Life Science, Leshan Normal University, Leshan, China
| | - Xiaoli Huang
- Shaoxing Testing Institute of Quality and Technical Supervision, Shaoxing, China
| | - Yang Pu
- Shaoxing Testing Institute of Quality and Technical Supervision, Shaoxing, China
| | - Guangfa Xie
- Zhejiang Collaborative Innovation Center for Full-Process Monitoring and Green Governance of Emerging Contaminants, College of Biology and Environmental Engineering, Zhejiang Shuren University, Hangzhou, China.
| | - Qi Peng
- National Engineering Research Center for Chinese CRW (branch center), School of Life and Environmental Sciences, Shaoxing University, Shaoxing, China.
| |
Collapse
|
281
|
Shang Y, Cao T, Ma X, Huang L, Wu M, Xu J, Wang J, Wang H, Wu S, Pandey V, Wu Z, Zhang W, Lobie PE, Han X, Zhu T. Estrogen-induced FXR1 promotes endocrine resistance and bone metastasis in breast cancer via BCL2 and GPX4. Front Cell Dev Biol 2025; 13:1563353. [PMID: 40196843 PMCID: PMC11973456 DOI: 10.3389/fcell.2025.1563353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 03/06/2025] [Indexed: 04/09/2025] Open
Abstract
Estrogen signaling dysregulation plays a critical role in the development of anti-estrogen resistance and bone metastasis of ER+ mammary carcinoma. Using quantitative proteomic screening, we identified FXR1 as an estrogen-regulated RNA-binding protein associated with anti-estrogen resistance. Mechanistically, estrogen and IGF1 facilitate FXR1 protein translation via the PI3K/AKT/mTOR/EIF4E pathway. FXR1 enhances cellular resistance to apoptosis and ferroptosis by facilitating the maturation of BCL2 pre-mRNA and stabilizing GPX4 mRNA, respectively. Anti-estrogen resistant cells exhibit elevated FXR1 expression, and FXR1 depletion restores their sensitivity to tamoxifen. Moreover, combining FXR1 depletion with a ferroptosis inducer induces synergistic lethal in anti-estrogen resistant cells. Finally, we provide proof-of-concept evidence supporting FXR1 antagonism as a potential treatment for bone metastases in ER+ breast cancer. Our findings highlight FXR1 as a promising therapeutic target to improve existing therapeutic regimes for ER+ breast cancer patients.
Collapse
Affiliation(s)
- Yinzhong Shang
- Department of Oncology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, National Key Laboratory of Immune Response and Immunotherapy, University of Science and Technology of China, Hefei, China
- Shenzhen Bay Laboratory, Institute of Biomedical Health Technology and Engineering, Shenzhen, China
| | - Tingfang Cao
- Department of Oncology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, National Key Laboratory of Immune Response and Immunotherapy, University of Science and Technology of China, Hefei, China
| | - Xin Ma
- Tsinghua Shenzhen International Graduate School, Institute of Biopharmaceutical and Health Engineering, Shenzhen, China
| | - Le Huang
- Department of Oncology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, National Key Laboratory of Immune Response and Immunotherapy, University of Science and Technology of China, Hefei, China
| | - Mingming Wu
- Department of Oncology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, National Key Laboratory of Immune Response and Immunotherapy, University of Science and Technology of China, Hefei, China
| | - Junchao Xu
- Department of Oncology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, National Key Laboratory of Immune Response and Immunotherapy, University of Science and Technology of China, Hefei, China
- Shenzhen Bay Laboratory, Institute of Biomedical Health Technology and Engineering, Shenzhen, China
| | - Jiarui Wang
- Department of Oncology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, National Key Laboratory of Immune Response and Immunotherapy, University of Science and Technology of China, Hefei, China
| | - Hao Wang
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Sheng Wu
- Department of Oncology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, National Key Laboratory of Immune Response and Immunotherapy, University of Science and Technology of China, Hefei, China
| | - Vijay Pandey
- Tsinghua Shenzhen International Graduate School, Institute of Biopharmaceutical and Health Engineering, Shenzhen, China
| | - Zhengsheng Wu
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Weijie Zhang
- Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Peter E. Lobie
- Shenzhen Bay Laboratory, Institute of Biomedical Health Technology and Engineering, Shenzhen, China
- Tsinghua Shenzhen International Graduate School, Institute of Biopharmaceutical and Health Engineering, Shenzhen, China
| | - Xinghua Han
- Department of Oncology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, National Key Laboratory of Immune Response and Immunotherapy, University of Science and Technology of China, Hefei, China
| | - Tao Zhu
- Department of Oncology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, National Key Laboratory of Immune Response and Immunotherapy, University of Science and Technology of China, Hefei, China
- Shenzhen Bay Laboratory, Institute of Biomedical Health Technology and Engineering, Shenzhen, China
- Tsinghua Shenzhen International Graduate School, Institute of Biopharmaceutical and Health Engineering, Shenzhen, China
- Anhui Key Laboratory of Molecular Oncology, Hefei, China
| |
Collapse
|
282
|
Khamineh Y, Ghiasvand M, Panahi-Alanagh S, Rastegarmand P, Zolghadri S, Stanek A. A Narrative Review of Quercetin's Role as a Bioactive Compound in Female Reproductive Disorders. Nutrients 2025; 17:1118. [PMID: 40218878 PMCID: PMC11990684 DOI: 10.3390/nu17071118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 04/14/2025] Open
Abstract
Lifestyle, nutrition, and metabolic status are central to maintaining women's reproductive health. With the rising prevalence of infertility, the need for effective strategies to preserve and enhance women's reproductive well-being has become increasingly urgent. Quercetin, a plant-derived polyphenol, has attracted growing interest for its potential to support reproductive health, regulate the hormonal balance, and improve fertility. This narrative review examines Quercetin's role in women's reproductive health and delineates its possible mechanisms of action in female reproductive system disorders, including polycystic ovary syndrome, recurrent miscarriage, and cervical, ovarian, and endometrial cancer (EC). By highlighting the biological pathways through which quercetin may exert its effects, this article underscores the need for further research and clinical trials to validate its therapeutic potential and its applications as a bioactive compound in foods aimed at enhancing women's reproductive health.
Collapse
Affiliation(s)
- Yasaman Khamineh
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran 1983969411, Iran; (Y.K.); (M.G.)
| | - Mahsa Ghiasvand
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran 1983969411, Iran; (Y.K.); (M.G.)
- Department of Stem Cells and Developmental Biology, Cell Science Research Centre, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran 3365166571, Iran
| | - Sanaz Panahi-Alanagh
- Department of Plant and Animal Biology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan 817467344, Iran;
| | - Parisa Rastegarmand
- Department of Microbiology, College of Science, Shiraz Branch, Islamic Azad University, Shiraz 1477893780, Iran;
| | - Samaneh Zolghadri
- Department of Biology, Jahrom Branch, Islamic Azad University, Jahrom 7414785318, Iran
| | - Agata Stanek
- Department of Internal Medicine, Metabolic Diseases and Angiology, Faculty of Health Sciences in Katowice, Medical University of Silesia, Ziolowa 45/47 St., 40-635 Katowice, Poland
| |
Collapse
|
283
|
Naoi M, Wu Y, Maruyama W, Shamoto-Nagai M. Phytochemicals Modulate Biosynthesis and Function of Serotonin, Dopamine, and Norepinephrine for Treatment of Monoamine Neurotransmission-Related Psychiatric Diseases. Int J Mol Sci 2025; 26:2916. [PMID: 40243512 PMCID: PMC11988947 DOI: 10.3390/ijms26072916] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 03/17/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025] Open
Abstract
Serotonin (5-HT), dopamine (DA), and norepinephrine (NE) are key monoamine neurotransmitters regulating behaviors, mood, and cognition. 5-HT affects early brain development, and its dysfunction induces brain vulnerability to stress, raising the risk of depression, anxiety, and autism in adulthood. These neurotransmitters are synthesized from tryptophan and tyrosine via hydroxylation and decarboxylation, and are metabolized by monoamine oxidase (MAO). This review aims to summarize the current findings on the role of dietary phytochemicals in modulating monoamine neurotransmitter biosynthesis, metabolism, and function, with an emphasis on their potential therapeutic applications in neuropsychiatric disorders. Phytochemicals exert antioxidant, neurotrophic, and neurohormonal activities, regulate gene expression, and induce epigenetic modifications. Phytoestrogens activate the estrogen receptors or estrogen-responsive elements of the promoter of target genes, enhance transcription of tryptophan hydroxylase and tyrosine hydroxylase, while inhibiting that of MAO. These compounds also influence the interaction between genetic and environmental factors, potentially reversing dysregulated neurotransmission and the brain architecture associated with neuropsychiatric conditions. Despite promising preclinical findings, clinical applications of phytochemicals remain challenging. Advances in nanotechnology and targeted delivery systems offer potential solutions to enhance clinical efficacy. This review discusses mechanisms, challenges, and strategies, underscoring the need for further research to advance phytochemical-based interventions for neuropsychiatric diseases.
Collapse
Affiliation(s)
- Makoto Naoi
- Department of Health and Nutritional Sciences, Faculty of Health Sciences, Aichi Gakuin University, 12 Araike, Iwasaki-cho, Nisshin 320-195, Aichi, Japan; (Y.W.); (W.M.); (M.S.-N.)
| | | | | | | |
Collapse
|
284
|
Zhou J, He M, Zhao Q, Shi E, Wang H, Ponkshe V, Song J, Wu Z, Ji D, Kranz G, Tscherne A, Schwenk-Zieger S, Razak NA, Hess J, Belka C, Zitzelsberger H, Ourailidis I, Stögbauer F, Boxberg M, Budczies J, Reichel CA, Canis M, Baumeister P, Wang H, Unger K, Mock A, Gires O. EGFR-mediated local invasiveness and response to Cetuximab in head and neck cancer. Mol Cancer 2025; 24:94. [PMID: 40121428 PMCID: PMC11929204 DOI: 10.1186/s12943-025-02290-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 03/04/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Recurrent/metastatic head and neck squamous cell carcinoma (R/M-HNSCC) is a severe, frequently lethal condition. Oncogene addiction to epidermal growth factor receptor (EGFR) is a hallmark of HNSCC, but the clinical efficacy of EGFR-targeted therapies remains low. Understanding molecular networks governing EGFR-driven progression is paramount to the exploration of (co)-treatment targets and predictive markers. METHODS We performed function-based mapping of differentially expressed genes in EGFR-mediated local invasion (fDEGs) using photoconvertible tracers and RNA-sequencing (RNA-seq) in a cellular 3D-model. RESULTS Upon alignment with public single-cell RNA-seq (scRNA-seq) datasets and HNSCC-specific regulons, a gene regulatory network of local invasion (invGRN) was inferred from gene expression data, which was overrepresented in budding tumors. InvGRN comprises the central hubs inhibin subunit beta alpha (INHBA) and snail family transcriptional repressor 2 (SNAI2), and druggable fDEGs integrin subunit beta 4 (ITGB4), laminin 5 (LAMB3/LAMC2), and sphingosine kinase 1 (SPHK1). Blockade of INHBA repressed local invasion and was reverted by activin A, laminin 5, and sphingosine-1-phosphate, demonstrating a functional interconnectivity of the invGRN. Epithelial-to-mesenchymal transition (EMT) of malignant cells and the invGRN are induced by newly defined EGFR-activity subtypes with prognostic value that are promoted by amphiregulin (AREG) and epiregulin (EREG). Importantly, co-inhibition of SPHK1 showed synthetic effects on Cetuximab-mediated invasion blockade and high expression of selected fDEGs was associated with response to Cetuximab in patient-derived xenotransplantation (PDX) and R/M-HNSCC patients. CONCLUSIONS We describe an actionable network of EGFR-mediated local invasion and define druggable effectors with predictive potential regarding the response of R/M-HNSCC to Cetuximab.
Collapse
Affiliation(s)
- Jiefu Zhou
- Department of Otorhinolaryngology, LMU University Hospital, LMU Munich, Munich, Germany
- Department of Sports Medicine, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, 410008, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Road 87, Changsha, 410008, China
- Hunan Engineering Research Center of Sports and Health, Changsha, 410008, China
| | - Min He
- Department of Otorhinolaryngology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Qiong Zhao
- Department of Otorhinolaryngology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Enxian Shi
- Department of Otorhinolaryngology, LMU University Hospital, LMU Munich, Munich, Germany
- Department of Dermatology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, Guangdong, China
| | - Hairong Wang
- Department of Otorhinolaryngology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Vaidehi Ponkshe
- Department of Otorhinolaryngology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Jiahang Song
- Department of Otorhinolaryngology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Zhengquan Wu
- Department of Otorhinolaryngology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Dongmei Ji
- Department of Medical Oncology, Department of Oncology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Gisela Kranz
- Department of Otorhinolaryngology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Anna Tscherne
- Department of Otorhinolaryngology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Sabina Schwenk-Zieger
- Department of Otorhinolaryngology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Nilofer Abdul Razak
- Department of Otorhinolaryngology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Julia Hess
- Research Unit Translational Metabolic Oncology, Institute for Diabetes and Cancer, Helmholtz Zentrum München, Deutsches Forschungszentrum Für Gesundheit Und Umwelt (GmbH), Neuherberg, Germany
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Claus Belka
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
- German Cancer Consortium (DKTK), Partner Site, Munich, Germany
- Comprehensive Cancer Center (CCC), Munich, Germany
| | - Horst Zitzelsberger
- Research Unit Translational Metabolic Oncology, Institute for Diabetes and Cancer, Helmholtz Zentrum München, Deutsches Forschungszentrum Für Gesundheit Und Umwelt (GmbH), Neuherberg, Germany
| | - Iordanis Ourailidis
- Institute of Pathology, University of Heidelberg, Im Neuenheimer Feld 224, 69120, Heidelberg, Germany
| | - Fabian Stögbauer
- Technical University of Munich, TUM School of Medicine and Health, Institute of General and Surgical Pathology, Munich, Germany
| | - Melanie Boxberg
- German Cancer Consortium (DKTK), Partner Site, Munich, Germany
| | - Jan Budczies
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Christoph A Reichel
- Department of Otorhinolaryngology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Martin Canis
- Department of Otorhinolaryngology, LMU University Hospital, LMU Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Philipp Baumeister
- Department of Otorhinolaryngology, LMU University Hospital, LMU Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Hongxia Wang
- Department of Medical Oncology, Department of Oncology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Kristian Unger
- Research Unit Translational Metabolic Oncology, Institute for Diabetes and Cancer, Helmholtz Zentrum München, Deutsches Forschungszentrum Für Gesundheit Und Umwelt (GmbH), Neuherberg, Germany
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
- German Cancer Consortium (DKTK), Partner Site, Munich, Germany
- Comprehensive Cancer Center (CCC), Munich, Germany
| | - Andreas Mock
- Institute of Pathology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Olivier Gires
- Department of Otorhinolaryngology, LMU University Hospital, LMU Munich, Munich, Germany.
| |
Collapse
|
285
|
Stanojević M, Sollner Dolenc M. Mechanisms of bisphenol A and its analogs as endocrine disruptors via nuclear receptors and related signaling pathways. Arch Toxicol 2025:10.1007/s00204-025-04025-z. [PMID: 40116906 DOI: 10.1007/s00204-025-04025-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 03/13/2025] [Indexed: 03/23/2025]
Abstract
Bisphenol A (BPA) is a widely used chemical that is slowly being phased out due to its toxic properties. The industry is therefore looking for alternatives in the form of BPA analogs. However, studies have shown that BPA analogs can have comparable or even stronger endocrine and toxic effects than BPA. This review describes various mechanisms and interactions of BPA analogs with individual nuclear receptors. They interfere with downstream signaling pathways not only by binding to the nuclear receptors, but also by various alternative mechanisms, such as altering receptor expression, affecting co-receptors, altering signal transduction pathways, and even epigenetic changes. Further studies are needed to fully investigate the potential synergistic and additive effects that may result. In the search for a less harmful alternative to BPA, affinity to the nuclear receptor may not be the decisive factor. We therefore recommend a different study approach to assess their effects on the endocrine system before new BPA analogs are introduced to the market to protect public health and the environment.
Collapse
Affiliation(s)
- Mark Stanojević
- Bisafe Doo, 1000, Ljubljana, Slovenia
- Faculty of Pharmacy, University of Ljubljana, 1000, Ljubljana, Slovenia
| | | |
Collapse
|
286
|
Ma JJ, Zhang LF, Xu DP, Wang Z, Meng YQ. Design, synthesis and antitumor activity of pentacyclic triterpenoid ursolic acid derivatives and oleanolic acid derivatives based on multi-target. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2025:1-18. [PMID: 40117208 DOI: 10.1080/10286020.2025.2473635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 02/24/2025] [Accepted: 02/24/2025] [Indexed: 03/23/2025]
Abstract
Totally twelve inhibitors of Survivin and Sp1 based on ursolic acid (UA) derivatives and oleanolic acid (OA) derivatives were designed and synthesized with modification at C-2, C-3 and C-28 of UA and OA. Their structures were confirmed by HRMS,1H NMR and 13C NMR. In vitro activity assay showed that these compounds can inhibit cell proliferation of HeLa, SKOV3, BGC-823 and HT1080 cells, especially compounds IV and X showed better inhibitory activity on these tumor cells than that of the positive control drug Gefitinib and similar to Vp-16. Mechanistically, selected compound may inhibit the proliferation of SKOV3 cells and trigger apoptosis by activating Sp1 to inhibit Survivin protein expression, which may be promising leading compounds for cancer therapy.
Collapse
Affiliation(s)
- Jun-Jiao Ma
- Department of Chemical Engineering, Shenyang University of Chemical Technology, Shenyang 110142, China
| | - Liang-Feng Zhang
- Department of Chemical Engineering, Shenyang University of Chemical Technology, Shenyang 110142, China
| | - Dong-Ping Xu
- Department of Chemical Engineering, Shenyang University of Chemical Technology, Shenyang 110142, China
| | - Zan Wang
- Department of Chemical Engineering, Shenyang University of Chemical Technology, Shenyang 110142, China
| | - Yan-Qiu Meng
- Department of Chemical Engineering, Shenyang University of Chemical Technology, Shenyang 110142, China
| |
Collapse
|
287
|
Luarte T, Hirmas-Olivares A, Gómez-Aburto V, Gómez K, Höfer J, Giesecke R, Přibylová PP, Martiník J, Pozo K, Chiang G, Pulgar J, Galbán-Malagón C. Bioaccumulation of PCBs and OCPs in Antarctic phytoplankton and zooplankton: Insights into bioconcentration and biomagnification in Fildes Bay. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 970:178986. [PMID: 40056554 DOI: 10.1016/j.scitotenv.2025.178986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 02/10/2025] [Accepted: 02/24/2025] [Indexed: 03/10/2025]
Abstract
This study was conducted at Fildes Bay, King George Island, Antarctica, from November 30th, 2019, to January 30th, 2020. We determined the concentrations of Organochlorine Pesticides (OCPs) and Polychlorinated Biphenyls (PCBs) in phytoplankton, and zooplankton. The most abundant compounds found were hexachlorobenzene (HCB) in phytoplankton and γ-HCH in zooplankton. We estimated the bioconcentration factors (BCFPHYTO) for phytoplankton and zooplankton (BCFZOO), as well as biomagnification factors (BMF) across trophic levels. Our findings indicate that BCF values were generally higher for compounds with a higher degree of chlorination, and noticeable biomagnification of these contaminants was observed, emphasizing the enhancement of contaminant concentrations from phytoplankton to zooplankton. This biomagnification varied depending on the specific compound and environmental conditions. Throughout the study period, the factors influencing bioconcentration and biomagnification were closely correlated with the seasonal dynamics of the phytoplankton biomass, which were affected by local conditions such as slower S and SE winds. These events enhanced the concentration of chlorophyll-a and POPs, influencing their transfer across trophic levels. Our study underscores the complex interplay between biotic and abiotic factors in determining the fate of these persistent pollutants in a rapidly changing polar environment. These findings highlight the need for continuing ongoing monitoring to understand the long-term impacts of environmental changes on the distribution and behavior of hazardous substances in remote regions like Antarctica.
Collapse
Affiliation(s)
- T Luarte
- Departamento de Ciencias Biológicas, Facultad Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile.
| | - A Hirmas-Olivares
- GEMA, Center for Genomics, Ecology & Environment, Universidad Mayor, Camino La Pirámide 5750, Huechuraba, Santiago, Chile
| | - V Gómez-Aburto
- GEMA, Center for Genomics, Ecology & Environment, Universidad Mayor, Camino La Pirámide 5750, Huechuraba, Santiago, Chile
| | - K Gómez
- Escuela de Ciencias del Mar, Pontifica Universidad Católica de Valparaíso, Chile
| | - J Höfer
- Escuela de Ciencias del Mar, Pontifica Universidad Católica de Valparaíso, Chile; Centro FONDAP de Investigación en Dinámica de Ecosistemas Marinos de Altas Latitudes (IDEAL), Valdivia, Chile
| | - R Giesecke
- Instituto de Ciencias Marinas y Limnológicas, Universidad Austral de Chile, Av. Rector Eduardo Morales Miranda 23, Valdivia, Chile; Centro FONDAP de Investigación en Dinámica de Ecosistemas Marinos de Altas Latitudes (IDEAL), Valdivia, Chile
| | - P P Přibylová
- RECETOX, Research Centre for Toxic Compounds in the Environment, Masaryk University, Kamenice 753/5, 625 00 Brno, Czech Republic
| | - J Martiník
- RECETOX, Research Centre for Toxic Compounds in the Environment, Masaryk University, Kamenice 753/5, 625 00 Brno, Czech Republic
| | - K Pozo
- RECETOX, Research Centre for Toxic Compounds in the Environment, Masaryk University, Kamenice 753/5, 625 00 Brno, Czech Republic; Facultad de Ingeniería y Tecnología, Universidad San Sebastián, Lientur 1457, Concepción, Bío Bío, Chile
| | - G Chiang
- Centro para la Resiliencia, Adaptación y Mitigación (CReAM), Universidad Mayor, Temuco, Chile; Centro de Investigación para la Sustentabilidad (CIS) & Departamento de Ecología y Biodiversidad, Facultad de Ciencias de la Vida, Universidad Nacional Andrés Bello, Santiago, Chile
| | - J Pulgar
- Departamento de Ecología y Biodiversidad, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - C Galbán-Malagón
- GEMA, Center for Genomics, Ecology & Environment, Universidad Mayor, Camino La Pirámide 5750, Huechuraba, Santiago, Chile; Dara Observatory Foundation, Santiago de Chile, Chile; Institute of Environment, Florida International University, University Park, Miami, FL 33199, USA.
| |
Collapse
|
288
|
Albeitawi S, Bani-Mousa SU, Jarrar B, Aloqaily I, Al-Shlool N, Alsheyab G, Kassab A, Qawasmi B, Awaisheh A. Associations Between Follicular Fluid Biomarkers and IVF/ICSI Outcomes in Normo-Ovulatory Women-A Systematic Review. Biomolecules 2025; 15:443. [PMID: 40149979 PMCID: PMC11940193 DOI: 10.3390/biom15030443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025] Open
Abstract
(1) Background: The follicular fluid (FF) comprises a large portion of ovarian follicles, and serves as both a communication and growth medium for oocytes, and thus should be representative of the metabolomic status of the follicle. This review aims to explore FF biomarkers as well as their effects on fertilization, oocyte, and embryo development, and later on implantation and maintenance of pregnancy. (2) Methods: This review was registered in the PROSPERO database with the ID: CRD42025633101. We parsed PubMed, Scopus, and Google Scholar for research on the effects of different FF biomarkers on IVF/ICSI outcomes in normo-ovulatory women. Included studies were assessed for risk of bias using the NOS scale. Data were extracted and tabulated by two independent researchers. (3) Results: 22 included articles, with a sample size range of 31 to 414 and a median of 60 participants, contained 61 biomarkers, including proteins, growth factors, steroid and polypeptide hormones, inflammation and oxidative stress markers, amino acids, vitamins, lipids of different types, and miRNAs. Most of the biomarkers studied had significant effects on IVF/ICSI outcomes, and seem to have roles in various cellular pathways responsible for oocyte and embryo growth, implantation, placental formation, and maintenance of pregnancy. The FF metabolome also seems to be interconnected, with its various components influencing the levels and activities of each other through feedback loops. (4) Conclusions: FF biomarkers can be utilized for diagnostic and therapeutic purposes in IVF; however, further studies are required for choosing the most promising ones due to heterogeneity of results. Widespread adoption of LC-MS and miRNA microarrays can help quantify a representative FF metabolome, and we see great potential for in vitro supplementation (IVS) of some FF biomarkers in improving IVF/ICSI outcomes.
Collapse
Affiliation(s)
- Soha Albeitawi
- Department of Pediatrics, Family Medicine and Obstetrics & Gynecology, Faculty of Medicine, Yarmouk University, Irbid 21163, Jordan; (B.J.); (I.A.); (N.A.-S.); (G.A.); (A.K.); (B.Q.); (A.A.)
| | | | - Baraa Jarrar
- Department of Pediatrics, Family Medicine and Obstetrics & Gynecology, Faculty of Medicine, Yarmouk University, Irbid 21163, Jordan; (B.J.); (I.A.); (N.A.-S.); (G.A.); (A.K.); (B.Q.); (A.A.)
| | - Ibrahim Aloqaily
- Department of Pediatrics, Family Medicine and Obstetrics & Gynecology, Faculty of Medicine, Yarmouk University, Irbid 21163, Jordan; (B.J.); (I.A.); (N.A.-S.); (G.A.); (A.K.); (B.Q.); (A.A.)
| | - Nour Al-Shlool
- Department of Pediatrics, Family Medicine and Obstetrics & Gynecology, Faculty of Medicine, Yarmouk University, Irbid 21163, Jordan; (B.J.); (I.A.); (N.A.-S.); (G.A.); (A.K.); (B.Q.); (A.A.)
| | - Ghaida Alsheyab
- Department of Pediatrics, Family Medicine and Obstetrics & Gynecology, Faculty of Medicine, Yarmouk University, Irbid 21163, Jordan; (B.J.); (I.A.); (N.A.-S.); (G.A.); (A.K.); (B.Q.); (A.A.)
| | - Ahmad Kassab
- Department of Pediatrics, Family Medicine and Obstetrics & Gynecology, Faculty of Medicine, Yarmouk University, Irbid 21163, Jordan; (B.J.); (I.A.); (N.A.-S.); (G.A.); (A.K.); (B.Q.); (A.A.)
| | - Baha’a Qawasmi
- Department of Pediatrics, Family Medicine and Obstetrics & Gynecology, Faculty of Medicine, Yarmouk University, Irbid 21163, Jordan; (B.J.); (I.A.); (N.A.-S.); (G.A.); (A.K.); (B.Q.); (A.A.)
| | - Abdalrahman Awaisheh
- Department of Pediatrics, Family Medicine and Obstetrics & Gynecology, Faculty of Medicine, Yarmouk University, Irbid 21163, Jordan; (B.J.); (I.A.); (N.A.-S.); (G.A.); (A.K.); (B.Q.); (A.A.)
| |
Collapse
|
289
|
de Santana MR, Argolo DS, Lima IS, dos Santos CC, Victor MM, Ramos GDS, do Nascimento RP, Ulrich H, Costa SL. Naringenin Exhibits Antiglioma Activity Related to Aryl Hydrocarbon Receptor Activity and IL-6, CCL2, and TNF-α Expression. Brain Sci 2025; 15:325. [PMID: 40149846 PMCID: PMC11940588 DOI: 10.3390/brainsci15030325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/13/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Glioblastoma (GBM) is a highly aggressive brain tumor characterized by rapid cell proliferation, invasive behavior, and chemoresistance. The aryl hydrocarbon receptor (AhR) is implicated in chemoresistance and immune evasion, making it a promising therapeutic target. Natural compounds such as flavonoids have gained attention for their anti-inflammatory, antioxidant, and anticancer properties. Among them, naringenin, a citrus-derived flavonoid, exerts antiproliferative, pro-apoptotic, and immunomodulatory effects. OBJECTIVES This study investigated the antiglioma effects of the flavonoid naringenin on the viability, growth, and migration of glioma cells and its potential role as an AhR modulator. METHODS Human (U87) and rat (C6) glioma cell lines were exposed to naringenin (10-300 µM) alone or in combination with the AhR agonist indole-3-carbinol (50 µM) for 24 to 48 h. Cell viability, scratch wound, and cell migration assays were performed. The expression of inflammatory markers was also analyzed by RT-qPCR. RESULTS Naringenin exerted dose- and time-dependent inhibition of cell viability and migration. The treatment decreased the gene expression of interleukin-6 (IL-6) and chemokine (CCL2), alongside increased tumor necrosis factor-alpha (TNF-α) expression, an effect reversed by the AhR agonist. CONCLUSIONS These findings highlight naringenin's potential as an antiglioma agent and its role in AhR signaling.
Collapse
Affiliation(s)
- Monique Reis de Santana
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Salvador 40231-300, Brazil; (M.R.d.S.); (D.S.A.); (I.S.L.); (C.C.d.S.); (R.P.d.N.)
| | - Deivison Silva Argolo
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Salvador 40231-300, Brazil; (M.R.d.S.); (D.S.A.); (I.S.L.); (C.C.d.S.); (R.P.d.N.)
| | - Irlã Santos Lima
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Salvador 40231-300, Brazil; (M.R.d.S.); (D.S.A.); (I.S.L.); (C.C.d.S.); (R.P.d.N.)
| | - Cleonice Creusa dos Santos
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Salvador 40231-300, Brazil; (M.R.d.S.); (D.S.A.); (I.S.L.); (C.C.d.S.); (R.P.d.N.)
| | - Maurício Moraes Victor
- Department of Organic Chemistry, Institute of Chemistry, Federal University of Bahia, Salvador 40231-300, Brazil; (M.M.V.); (G.d.S.R.)
| | - Gabriel dos Santos Ramos
- Department of Organic Chemistry, Institute of Chemistry, Federal University of Bahia, Salvador 40231-300, Brazil; (M.M.V.); (G.d.S.R.)
| | - Ravena Pereira do Nascimento
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Salvador 40231-300, Brazil; (M.R.d.S.); (D.S.A.); (I.S.L.); (C.C.d.S.); (R.P.d.N.)
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-220, Brazil;
| | - Silvia Lima Costa
- Laboratory of Neurochemistry and Cellular Biology, Institute of Health Sciences, Federal University of Bahia, Salvador 40231-300, Brazil; (M.R.d.S.); (D.S.A.); (I.S.L.); (C.C.d.S.); (R.P.d.N.)
- National Institute of Translational Neuroscience (INNT), Rio de Janeiro 21941-902, Brazil
| |
Collapse
|
290
|
Uldbjerg CS, Rantakokko P, Lim YH, Petersen JH, Sørensen KM, Coull BA, Lindh C, Hauser R, Bräuner EV, Skakkebæk NE, Priskorn L, Juul A. Prenatal exposure to organochlorine pesticides and polychlorinated biphenyls and risk of testicular germ cell cancer later in life. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 970:179054. [PMID: 40056550 DOI: 10.1016/j.scitotenv.2025.179054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 01/31/2025] [Accepted: 03/03/2025] [Indexed: 03/10/2025]
Abstract
INTRODUCTION Exposure to environmental chemicals during fetal development may increase the risk of testicular germ cell cancer (TGCC), but few studies have tested the hypothesis. We focused on organochlorine pesticides (OCPs) and polychlorinated biphenyls (PCBs), previously investigated in relation to other male reproductive health outcomes. METHODS We conducted a nested case-control study of 332 mother-son pairs, comprising 65 TGCC cases and 267 controls, identified from a Danish Pregnancy Screening Registry with biobanked serum samples collected from pregnant women in 1985-1995, when exposure to the studied chemicals was relatively high. We quantified seven OCPs and 13 PCB congeners in maternal serum by gas chromatography tandem mass spectrometry. TGCC diagnoses and covariate information were derived from the nationwide Danish registries. We estimated associations between individual chemicals and their mixture with the risk of TGCC through adapted Cox regression and quantile g-computation models. RESULTS Median age at TGCC diagnosis was 24.7 years. In main analyses, associations between individual OCPs and PCBs and risk of TGCC showed either slightly higher risks or no association (close to Hazard Ratios (HR) of 1.00), with confidence intervals overlapping unity. In mixture analyses, simultaneously increasing all chemical concentrations by one quartile resulted in a slightly higher risk of TGCC (HR 1.11, 95 % CI: 0.61; 2.05) after adjusting for confounders. Sensitivity analyses investigating tertiles of concentrations did not change the overall pattern of results. CONCLUSIONS Prenatal exposure to OCPs and PCBs, quantified by concentrations in maternal pregnancy serum, was not associated with later risk of TGCC.
Collapse
Affiliation(s)
- Cecilie S Uldbjerg
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, University of Copenhagen, Denmark; International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark.
| | - Panu Rantakokko
- Department of Health Security, Finnish Institute for Health and Welfare, 70701 Kuopio, Finland
| | - Youn-Hee Lim
- Section of Environmental Health, Department of Public Health, University of Copenhagen, Copenhagen, Denmark; Department of Preventive Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jørgen H Petersen
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, University of Copenhagen, Denmark; International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; Section of Biostatistics, Department of Public Health, University of Copenhagen, Denmark
| | | | - Brent A Coull
- Department of Biostatistics, T.H. Chan School of Public Health, Harvard University, USA
| | - Christian Lindh
- Division of Occupational and Environmental Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Russ Hauser
- Department of Environmental Health, T.H. Chan School of Public Health, Harvard University, USA
| | - Elvira V Bräuner
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, University of Copenhagen, Denmark; International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Niels E Skakkebæk
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, University of Copenhagen, Denmark; International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Lærke Priskorn
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, University of Copenhagen, Denmark; International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Anders Juul
- Department of Growth and Reproduction, Copenhagen University Hospital - Rigshospitalet, University of Copenhagen, Denmark; International Centre for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
291
|
Al Hashami ZS, van der Vegt B, Mourits MJ, Kluiver J, van den Berg A. miRNA-dependent resistance mechanisms to anti-hormonal therapies in estrogen receptor-positive breast cancer patients. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200941. [PMID: 40190354 PMCID: PMC11969448 DOI: 10.1016/j.omton.2025.200941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
The estrogen receptor (ERα) is expressed in 70%-80% of breast cancers and is a target of endocrine therapy. However, resistance to endocrine therapy poses a significant clinical challenge. MicroRNAs (miRNAs) have emerged as critical players in oncogenesis and as modulators of therapy response. This review provides an overview of miRNAs that modulate anti-hormonal drug responses. We identified 56 miRNAs associated with resistance to endocrine therapy. These miRNAs had a total of 40 proven target genes that were grouped based on their function under currently known resistance mechanisms, including ER modulation, signaling pathway activation, cell-cycle modulation, and other mechanisms. For a limited number of miRNA-target gene interactions, the relevance of the identified target gene(s) was confirmed by copy or rescue of the miRNA-induced phenotype. Overall, this review highlights critical roles of miRNAs as crucial mediators of resistance to anti-hormonal therapy. The identified miRNA-target gene interactions can serve as a foundation for future functional studies exploring the potential of selected miRNAs in overcoming drug resistance, which might improve outcomes for breast cancer patients.
Collapse
Affiliation(s)
- Zainab Salam Al Hashami
- Department of Pathology and Medical Biology, University of Groningen, Groningen, the Netherlands
- University Medical Centre Groningen, Groningen, the Netherlands
- Department of Biochemistry, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Bert van der Vegt
- Department of Pathology and Medical Biology, University of Groningen, Groningen, the Netherlands
- University Medical Centre Groningen, Groningen, the Netherlands
| | - Marian J.E. Mourits
- Department of Gynaecological Oncology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Joost Kluiver
- Department of Pathology and Medical Biology, University of Groningen, Groningen, the Netherlands
- University Medical Centre Groningen, Groningen, the Netherlands
| | - Anke van den Berg
- Department of Pathology and Medical Biology, University of Groningen, Groningen, the Netherlands
- University Medical Centre Groningen, Groningen, the Netherlands
| |
Collapse
|
292
|
Duplan E, Bernardin A, Goiran T, Leroudier N, Casimiro M, Pestell R, Tanaka S, Malleval C, Honnorat J, Idbaih A, Martin L, Castel H, Checler F, Alves da Costa C. α-synuclein expression in glioblastoma restores tumor suppressor function and rescues temozolomide drug resistance. Cell Death Dis 2025; 16:188. [PMID: 40108111 PMCID: PMC11923286 DOI: 10.1038/s41419-025-07509-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 02/17/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025]
Abstract
Several studies have shown that Parkinson's disease causative gene products, including α-synuclein (α-syn), display tight links with the tumor suppressor p53. The purpose of this study is to determine the implication of α-syn in glioblastoma development and elucidate how it elicits a tumor suppressor function. We show that the expression of α-syn, a TP53 transcriptional target and a key molecular player in Parkinson's disease, is detected in 1p/19q-codeleted and isocitrate dehydrogenase (IDH)-mutant oligodendroglioma and in IDH-wild-type glioblastoma, while reduced in glioblastoma biopsies, corroborating the link of α-syn expression with a better prognosis among all glioma patients. Accordingly, protein expression is drastically reduced in oligodendrogliomas and glioblastoma biopsies. This could be accounted for by a reduction of p53 transcriptional activity in these samples. Interestingly, genetic manipulation of p53 in glioblastoma cells and in mouse brain shows that p53 up-regulates α-synuclein, a phenotype fully abolished by the prominent p53 hot spot mutation R175H. Downstream to its p53-linked control, α-syn lowers cyclin D1 protein and mRNA levels and reduces glioblastoma cells proliferation in a cyclin D1-dependent-manner. Further, in temozolomide (TMZ)-resistant U87 cells, α-syn reduces O6-methylguanine-DNA methyltransferase (MGMT) expression and rescues drug sensitivity by a mechanism implying its transcriptional activation by X-box binding protein 1 (XBP1), an effector of the UPR response. Furthermore, α-syn lowers MGMT and cyclin D1 (CCDN1) expressions and reduces tumor development in allografted mice. Overall, our data reveals a new role of α-syn as an oligodendroglioma biomarker and as a glioblastoma tumor suppressor capable of either potentiate TMZ effect or avoid TMZ-associated resistance.
Collapse
Affiliation(s)
- Eric Duplan
- University Côte d'azur, INSERM U1323, CNRS UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), team labeled "Laboratory of Excellence (LABEX) Distalz", 06560, Valbonne, France.
| | - Aurore Bernardin
- University Côte d'azur, INSERM U1323, CNRS UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), team labeled "Laboratory of Excellence (LABEX) Distalz", 06560, Valbonne, France
| | - Thomas Goiran
- University Côte d'azur, INSERM U1323, CNRS UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), team labeled "Laboratory of Excellence (LABEX) Distalz", 06560, Valbonne, France
| | - Nathalie Leroudier
- University Côte d'azur, INSERM U1323, CNRS UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), team labeled "Laboratory of Excellence (LABEX) Distalz", 06560, Valbonne, France
| | - Mathew Casimiro
- Department of Science and Mathematics, Abraham Baldwin Agricultural College, Tifton, GA, 31794, USA
| | - Richard Pestell
- Pennsylvania Cancer and Regenerative Medicine Research Center, Baruch S. Blumberg Institute, Pennsylvania Biotechnology Center, Wynnewood, PA, 19096, USA
- The Wistar Institute, Philadelphia, PA, 19107, USA
- Garvan Institute of Medical Research, and, St Vincent's Clinical School, UNSW Sydney, 384 Victoria Street, Darlinghurst, NSW, 2010, Australia
| | - Shinya Tanaka
- Department of Cancer Pathology, Faculty of Medicine, and Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, N15, W7, Sapporo, 060-8638, Japan
| | - Celine Malleval
- Department of Neuro-Oncology, Hospices Civils de Lyon, Hôpital Neurologique, Institute MeLiS-UCBL-CNRS UMR 5284. INSERM U1314, University Claude Bernard Lyon 1, Lyon, 69008, France
| | - Jerome Honnorat
- Department of Neuro-Oncology, Hospices Civils de Lyon, Hôpital Neurologique, Institute MeLiS-UCBL-CNRS UMR 5284. INSERM U1314, University Claude Bernard Lyon 1, Lyon, 69008, France
| | - Ahmed Idbaih
- Sorbonne Université, AP-HP, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, DMU Neurosciences, Service de Neuro-Oncologie-Institut de Neurologie, F-75013, Paris, France
| | - Lucie Martin
- Univ Rouen Normandie, Inserm U1245, Normandie Univ, F-76000, Rouen, France
- Institute of Research and Innovation in Biomedicine (IRIB), 76000, Rouen, France
| | - Hélène Castel
- Univ Rouen Normandie, Inserm U1245, Normandie Univ, F-76000, Rouen, France
- Institute of Research and Innovation in Biomedicine (IRIB), 76000, Rouen, France
- Cancer and Cognition Platform, Normandie Univ, 14000, Caen, France
| | - Frédéric Checler
- University Côte d'azur, INSERM U1323, CNRS UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), team labeled "Laboratory of Excellence (LABEX) Distalz", 06560, Valbonne, France
| | - Cristine Alves da Costa
- University Côte d'azur, INSERM U1323, CNRS UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), team labeled "Laboratory of Excellence (LABEX) Distalz", 06560, Valbonne, France.
| |
Collapse
|
293
|
Yu X, He Y, Kamenecka TM, Kojetin DJ. Towards a unified molecular mechanism for liganddependent activation of NR4A-RXR heterodimers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.19.642122. [PMID: 40166180 PMCID: PMC11956975 DOI: 10.1101/2025.03.19.642122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
A subset of nuclear receptors (NRs) function as permissive heterodimers with retinoid X receptor (RXR), defined by transcriptional activation in response to binding RXR agonist ligands. Permissive NR-RXR activation operates via a classical pharmacological mechanism, where binding of an RXR agonist increases coactivator recruitment to the heterodimer. However, we previously demonstrated that transcriptional activation of permissive Nurr1-RXRα (NR4A2-NR2B1) heterodimers by an RXR ligand set, which included pharmacological RXR agonists and selective Nurr1-RXRα agonists that function as antagonists of RXRα homodimers, occurs via a non-classical mechanism: ligand-binding domain (LBD) heterodimer dissociation (Yu et al., 2023). Here, we extend mechanistic ligand profiling of the same RXR ligand set to Nur77-RXRγ (NR4A1-NR2B3), which is evolutionarily related to Nurr1-RXRα. Biochemical and NMR protein-protein interaction profiling along with cellular transcription studies indicate that the RXR ligand set, which lacks selective Nur77-RXRγ agonists, may influence Nur77-RXRγ transcriptional activation through both classical pharmacological activation and LBD heterodimer dissociation. However, upon reanalyzing our previously published data for Nurr1-RXRα, we found that the inclusion of selective Nurr1-RXRα agonists was essential for elucidating the LBD heterodimer dissociation mechanism. Our findings underscore the need for a more functionally diverse RXR ligand set to explore Nur77-RXRγ activation and unify LBD heterodimer dissociation as a potential targeting mechanism for NR4A-RXR heterodimers in neurodegenerative and inflammatory diseases.
Collapse
Affiliation(s)
- Xiaoyu Yu
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, United States
- Department of Integrative Structural and Computational Biology, Scripps Research and The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, Florida, United States
| | - Yuanjun He
- Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, United States
| | - Theodore M. Kamenecka
- Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, United States
| | - Douglas J. Kojetin
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, United States
- Department of Integrative Structural and Computational Biology, Scripps Research and The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, Florida, United States
- Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, United States
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, United States
- Center for Applied AI in Protein Dynamics, Vanderbilt University, Nashville, Tennessee, United States
| |
Collapse
|
294
|
Georgiev P, Belka M, Ulenberg S, Kroll D, Marciniak B, Drążkowska I, Bączek T, Płotka-Wasylka J. Headspace Extraction onto a 3D-Printed Device for GC-MS Quantification of Polychlorinated Biphenyls in Newborn Urine. Int J Mol Sci 2025; 26:2755. [PMID: 40141397 PMCID: PMC11942790 DOI: 10.3390/ijms26062755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/11/2025] [Accepted: 03/14/2025] [Indexed: 03/28/2025] Open
Abstract
Polychlorinated biphenyls (PCBs) are persistent organic pollutants that pose significant health risks, especially for neonates. Traditional urine analysis methods for PCBs are often complex and prone to contamination. This study introduces a novel, efficient, and contamination-free method for PCB analysis in neonatal urine using 3D-printed extraction devices. A headspace extraction method was developed, utilizing a 3D-printed device containing C18-modified silica particles. Urine samples were heated to 90 °C, and volatile PCBs were sorbed onto the particles. The method was optimized for maximum extraction efficiency and selectivity, demonstrating excellent linearity, precision, and accuracy. The optimized method was successfully applied to analyze neonatal urine samples, revealing detectable levels of PCBs. This innovative approach, leveraging 3D-printed devices, offers a promising solution for sample preparation, minimizing contamination risks and enabling the analysis of volatile compounds. The customizable nature of 3D-printed devices opens up possibilities for future advancements in environmental analysis.
Collapse
Affiliation(s)
- Paweł Georgiev
- Department of Pharmaceutical Chemistry, Medical University of Gdańsk, J. Hallera 107, 80-416 Gdańsk, Poland; (P.G.)
| | - Mariusz Belka
- Department of Pharmaceutical Chemistry, Medical University of Gdańsk, J. Hallera 107, 80-416 Gdańsk, Poland; (P.G.)
| | - Szymon Ulenberg
- Department of Pharmaceutical Chemistry, Medical University of Gdańsk, J. Hallera 107, 80-416 Gdańsk, Poland; (P.G.)
| | - Dagmara Kroll
- Department of Pharmaceutical Chemistry, Medical University of Gdańsk, J. Hallera 107, 80-416 Gdańsk, Poland; (P.G.)
| | - Bartosz Marciniak
- Department of Pharmaceutical Chemistry, Medical University of Gdańsk, J. Hallera 107, 80-416 Gdańsk, Poland; (P.G.)
| | - Izabela Drążkowska
- Division of Neonatology, Medical University of Gdańsk, 80-210 Gdańsk, Poland
| | - Tomasz Bączek
- Department of Pharmaceutical Chemistry, Medical University of Gdańsk, J. Hallera 107, 80-416 Gdańsk, Poland; (P.G.)
| | - Justyna Płotka-Wasylka
- Department of Analytical Chemistry, Faculty of Chemistry, Gdańsk University of Technology, G. Narutowicza 11/12, 80-233 Gdańsk, Poland;
- BioTechMed Center, Gdańsk University of Technology, G. Narutowicza 11/12, 80-233 Gdańsk, Poland
| |
Collapse
|
295
|
Halabitska I, Petakh P, Kamyshnyi O. Metformin as a disease-modifying therapy in osteoarthritis: bridging metabolism and joint health. Front Pharmacol 2025; 16:1567544. [PMID: 40176893 PMCID: PMC11962732 DOI: 10.3389/fphar.2025.1567544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 02/27/2025] [Indexed: 04/05/2025] Open
Abstract
Background Osteoarthritis (OA) and impaired glucose tolerance (IGT) frequently coexist, leading to compounded clinical and metabolic challenges. This study investigates the effects of metformin in improving both clinical outcomes (pain, stiffness, physical function) and metabolic parameters (inflammatory markers, lipid profile, BMI) in patients with knee OA and IGT. Methods The study included 60 patients diagnosed with knee OA and IGT. Participants were divided into two groups: 26 patients received standard OA treatment without metformin (Without Metf), while 34 received metformin (500 mg twice daily) for 3 months, in addition to standard treatment (With Metf). Clinical assessments (WOMAC, Lequesne Algofunctional Index, KOOS, VAS) and metabolic markers (CRP, NLR, SOD, lipid profile, BMI) were measured before treatment, after 1 month, and after 3 months. Results The With Metf group showed significantly greater improvements in pain, stiffness, physical function, and quality of life compared to the Without Metf group. Metformin also led to significant reductions in inflammatory markers and improvements in lipid profiles and metabolic health indicators. The With Metf group demonstrated enhanced BMI, waist-to-hip ratio, and waist-to-height ratio. Furthermore, the need for increased NSAID doses was predicted by factors such as pain severity and inflammatory markers. Conclusion Metformin effectively alleviates osteoarthritis symptoms and improves metabolic health in patients with both OA and IGT. Further research is needed to explore its long-term effects on joint health, inflammatory markers, and its potential role in OA management in patients without IGT.
Collapse
Affiliation(s)
- Iryna Halabitska
- Department of Therapy and Family Medicine, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Pavlo Petakh
- Department of Biochemistry and Pharmacology, Uzhhorod National University, Uzhhorod, Ukraine
| | - Oleksandr Kamyshnyi
- Department of Microbiology, Virology, and Immunology, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| |
Collapse
|
296
|
Black G, Anderson BN, Wong L, Alaimo CP, He G, Denison MS, Bennett DH, Tancredi D, Durbin-Johnson B, Hammock BD, Chowdhary P, Rubin R, Young TM. Comprehensive Nontargeted Analysis of Drinking Water Supplies to Identify Chemicals Associated with Estrogen Receptor Agonism or Present in Regions of Elevated Breast Cancer Occurrence. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:5237-5248. [PMID: 40042489 PMCID: PMC11924233 DOI: 10.1021/acs.est.4c12204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/19/2025]
Abstract
To explore the hypothesis that differential exposures to estrogen active chemicals may contribute to regional disparities in cancer incidence, a comprehensive targeted and nontargeted analysis was conducted over two seasons (2020) for drinking water samples from 120 households served by 8 public water systems (4 with historically elevated breast cancer incidence) and from 15 brands of retail water. All samples were analyzed using gas and liquid chromatography with high-resolution mass spectrometry and a bioassay for estrogen receptor agonism. Target compounds included disinfection byproducts, per- and polyfluoroalkyl substances (PFAS), trace elements, and compounds selected for their possible relation to breast cancer. Over 7500 GC and LC nontargeted molecular features passed all quality control filters in each sampling season and were prioritized for identification if they were related to measured estrogen receptor agonism or were present at higher levels in areas with high breast cancer incidence (n = 1036). Benzothiazole-2-sulfonic acid, acetyl tributyl citrate, and diphenyl sulfone were among the prioritized and confirmed nontarget compounds. Nine polycyclic aromatic hydrocarbons and two ketone derivatives displayed significant negative correlations with estrogen receptor agonism. Many prioritized compounds remained unidentified, as 84.4% of the LC features and 77.5% of the GC features could not be annotated with high confidence.
Collapse
Affiliation(s)
- Gabrielle
P. Black
- Department
of Civil and Environmental Engineering, University of California, Davis One Shields Ave., Davis, California 95616, United States
| | - Berkley N. Anderson
- Department
of Civil and Environmental Engineering, University of California, Davis One Shields Ave., Davis, California 95616, United States
| | - Luann Wong
- Department
of Civil and Environmental Engineering, University of California, Davis One Shields Ave., Davis, California 95616, United States
| | - Christopher P. Alaimo
- Department
of Civil and Environmental Engineering, University of California, Davis One Shields Ave., Davis, California 95616, United States
| | - Guochun He
- Department
of Environmental Toxicology, University
of California, Davis
One Shields Ave., Davis, California 95616, United States
| | - Michael S. Denison
- Department
of Environmental Toxicology, University
of California, Davis
One Shields Ave., Davis, California 95616, United States
| | - Deborah H. Bennett
- Department
of Public Health Sciences, University of
California, Davis One
Shields Ave., Davis, California 95616, United States
| | - Daniel Tancredi
- Department
of Public Health Sciences, University of
California, Davis One
Shields Ave., Davis, California 95616, United States
| | - Blythe Durbin-Johnson
- Department
of Public Health Sciences, University of
California, Davis One
Shields Ave., Davis, California 95616, United States
| | - Bruce D. Hammock
- Department
of Entomology and Cancer Center, University
of California, Davis
One Shields Ave., Davis, California 95616, United States
| | - Pujeeta Chowdhary
- Breast
Cancer Prevention Partners, 1388 Sutter St #901, San Francisco, California 94109, United States
| | - Rainbow Rubin
- Breast
Cancer Prevention Partners, 1388 Sutter St #901, San Francisco, California 94109, United States
| | - Thomas M. Young
- Department
of Civil and Environmental Engineering, University of California, Davis One Shields Ave., Davis, California 95616, United States
| |
Collapse
|
297
|
Minacori M, Fiorini S, Perugini M, Iannetta A, Meschiari G, Chichiarelli S, Altieri F, Natali PG, Eufemi M. AhR and STAT3: A Dangerous Duo in Chemical Carcinogenesis. Int J Mol Sci 2025; 26:2744. [PMID: 40141386 PMCID: PMC11943011 DOI: 10.3390/ijms26062744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/13/2025] [Accepted: 03/15/2025] [Indexed: 03/28/2025] Open
Abstract
Human chemical carcinogenesis is a multistage process where chemicals or their metabolites cause irreversible changes in normal cell physiology, eventually leading to uncontrolled proliferation, transforming a normal cell into a cancerous one. Signal transducer and activator of transcription 3 (STAT3) is a cytoplasmic transcription factor that regulates cell proliferation, differentiation, apoptosis, angiogenesis, inflammation, and immune responses. Its aberrant activation triggers tumor progression by promoting the expression of oncogenic genes; thus, STAT3 is classified as an oncoprotein. The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that responds to a wide variety of chemicals, including carcinogens like dioxins, inducing genes associated with detoxification, proliferation, and immune regulation. Recent reports show that AhR plays a critical role in cancer development and maintenance. AhR may interact with signaling pathways, like the STAT3 pathway, which mediates the carcinogenic effects of several pollutants. Various chemical agents, such as industrial waste and hydrocarbon compounds, can alter the expression or signaling activity of AhR and STAT3 pathways, leading to different types of cancers. Understanding the complex STAT3-AhR network in the regulation of chemical carcinogenesis could open new avenues for cancer prevention or treatment, particularly in personalized medicine, aiming to improve life expectancy and achieving a complete cure.
Collapse
Affiliation(s)
- Marco Minacori
- Department of Biochemical Science “A. Rossi Fanelli”, Faculty of Pharmacy and Medicine, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy; (M.M.); (S.F.); (G.M.); (S.C.); (F.A.); (M.E.)
- Department of Bioscience and Agro-Food and Environmental Technology, University of Teramo, Campus “Aurelio Saliceti”, Via R. Balzarini 1, 64100 Teramo, Italy; (M.P.); (A.I.)
| | - Sara Fiorini
- Department of Biochemical Science “A. Rossi Fanelli”, Faculty of Pharmacy and Medicine, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy; (M.M.); (S.F.); (G.M.); (S.C.); (F.A.); (M.E.)
| | - Monia Perugini
- Department of Bioscience and Agro-Food and Environmental Technology, University of Teramo, Campus “Aurelio Saliceti”, Via R. Balzarini 1, 64100 Teramo, Italy; (M.P.); (A.I.)
| | - Annamaria Iannetta
- Department of Bioscience and Agro-Food and Environmental Technology, University of Teramo, Campus “Aurelio Saliceti”, Via R. Balzarini 1, 64100 Teramo, Italy; (M.P.); (A.I.)
| | - Giorgia Meschiari
- Department of Biochemical Science “A. Rossi Fanelli”, Faculty of Pharmacy and Medicine, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy; (M.M.); (S.F.); (G.M.); (S.C.); (F.A.); (M.E.)
| | - Silvia Chichiarelli
- Department of Biochemical Science “A. Rossi Fanelli”, Faculty of Pharmacy and Medicine, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy; (M.M.); (S.F.); (G.M.); (S.C.); (F.A.); (M.E.)
| | - Fabio Altieri
- Department of Biochemical Science “A. Rossi Fanelli”, Faculty of Pharmacy and Medicine, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy; (M.M.); (S.F.); (G.M.); (S.C.); (F.A.); (M.E.)
| | - Pier Giorgio Natali
- Collegium Ramazzini, Castello di Bentivoglio, Via Saliceto, 3, 40010 Bologna, Italy
| | - Margherita Eufemi
- Department of Biochemical Science “A. Rossi Fanelli”, Faculty of Pharmacy and Medicine, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy; (M.M.); (S.F.); (G.M.); (S.C.); (F.A.); (M.E.)
| |
Collapse
|
298
|
Kopalli SR, Behl T, Kyada A, Rekha MM, Kundlas M, Rani P, Nathiya D, Satyam Naidu K, Gulati M, Bhise M, Gupta P, Wal P, Fareed M, Ramniwas S, Koppula S, Gasmi A. Synaptic plasticity and neuroprotection: The molecular impact of flavonoids on neurodegenerative disease progression. Neuroscience 2025; 569:161-183. [PMID: 39922366 DOI: 10.1016/j.neuroscience.2025.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/11/2025] [Accepted: 02/03/2025] [Indexed: 02/10/2025]
Abstract
Flavonoids are a broad family of polyphenolic chemicals that are present in a wide variety of fruits, vegetables, and medicinal plants. Because of their neuroprotective qualities, flavonoids have attracted a lot of interest. The potential of flavonoids to control synaptic plasticity-a crucial process underlying memory, learning, and cognitive function-is becoming more and more clear. Dysregulation of synaptic plasticity is a feature of neurodegenerative diseases such as amyotrophic lateral sclerosis (0.4 %), Parkinson's (1-2 %), Alzheimer's (5-7 %), and Huntington's ((0.2 %)). This review discusses the molecular mechanisms via which flavonoids influence synaptic plasticity as well as their therapeutic potential in neurodegenerative diseases. Flavonoids modulate key signaling pathways such as MAPK/ERK and PI3K/Akt/mTOR to support neuroprotection, synaptic plasticity, and neuronal health, while also influencing neurotrophic factors (BDNF, NGF) and their receptors (TrkB, TrkA). They regulate neurotransmitter receptors like GABA, AMPA, and NMDA to balance excitatory and inhibitory transmission, and exert antioxidant effects via the Nrf2-ARE pathway and anti-inflammatory actions by inhibiting NF-κB signaling, highlighting their potential for treating neurodegenerative diseases. These varied reactions support the preservation of synapse function and neuronal integrity in the face of neurodegenerative insults. Flavonoids can reduce the symptoms of neurodegeneration, prevent synaptic loss, and enhance cognitive function, according to experimental studies. However, there are still obstacles to using these findings in clinical settings, such as limited bioavailability and the need for consistent dose. The focus of future research should be on improving flavonoid delivery systems and combining them with conventional medications.
Collapse
Affiliation(s)
- Spandana Rajendra Kopalli
- Department of Bioscience and Biotechnology, Sejong University, Gwangjin-gu, Seoul 05006 Republic of Korea
| | - Tapan Behl
- Amity School of Pharmaceutical Sciences, Amity University, Punjab 140306, India
| | - Ashishkumar Kyada
- Marwadi University Research Center, Department of Pharmaceutical Sciences, Faculty of Health Sciences, Marwadi University, Rajkot 360003 Gujarat, India
| | - M M Rekha
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Mayank Kundlas
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura 140401 Punjab, India
| | - Pooja Rani
- Department of Pharmacy, Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307 Punjab, India
| | - Deepak Nathiya
- Department of Pharmacy Practice, NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - K Satyam Naidu
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 1444411, India; ARCCIM, Faculty of Health, University of Technology Sydney, Ultimo, NSW 20227, Australia
| | | | | | - Pranay Wal
- PSIT- Pranveer Singh Institute of Technology, Pharmacy Kanpur UP, India
| | - Mohammad Fareed
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh 13713, Saudi Arabia
| | - Seema Ramniwas
- University Centre for Research and Development, Department of Biotechnology, Chandigarh University, Gharuan, Mohali 140413 Punjab, India
| | - Sushruta Koppula
- College of Biomedical and Health Sciences, Konkuk University, Chungju-Si, Chungcheongbuk Do 27478, Republic of Korea.
| | - Amin Gasmi
- Societe Francophone de Nutritherapie et de Nutrigenetique Appliquee, Villeurbanne, France; International Institute of Nutrition and Micronutrient Sciences, Saint-Etienne, France
| |
Collapse
|
299
|
Villagrán-Silva F, Loren P, Sandoval C, Lanas F, Salazar LA. Circulating microRNAs as Potential Biomarkers of Overweight and Obesity in Adults: A Narrative Review. Genes (Basel) 2025; 16:349. [PMID: 40149500 PMCID: PMC11942292 DOI: 10.3390/genes16030349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/29/2025] Open
Abstract
In an obesogenic environment, such as the one we have been experiencing in recent decades, epigenetics provides answers to the relationship between hereditary and environmentally acquired patterns that have significantly contributed to the global rise in obesity prevalence. MicroRNA (miRNA) constitutes a diminutive non-coding small RNA molecule, 20 to 24 nucleotides in length, that functions as a regulator of gene regulation at the post-translational level. Circulating miRNAs (c-miRNAs) have been detected in multiple body fluids, including blood, plasma, serum, saliva, milk from breastfeeding mothers, and urine. These molecules hold significant therapeutic value and serve as extracellular biomarkers in metabolic diseases. They aid in the diagnosis and tracking of therapy responses, as well as dietary and physical habit modifications. Researchers have studied c-miRNAs as potential biomarkers for diagnosing and characterizing systemic diseases in people of all ages and backgrounds since then. These conditions encompass dyslipidemia, type 2 diabetes mellitus (T2DM), cardiovascular risk, metabolic syndrome, cardiovascular diseases, and obesity. This review therefore analyzes the usefulness of c-miRNAs as therapeutic markers over the past decades. It also provides an update on c-miRNAs associated with general obesity and overweight, as well as with the most prevalent pathologies in the adult population. It also examines the effect of different nutritional approaches and physical activity regarding the activity of miRNAs in circulation in adults with overweight or general obesity. All of this is done with the aim of evaluating their potential use as biomarkers in various research contexts related to overweight and obesity in adults.
Collapse
Affiliation(s)
- Francisca Villagrán-Silva
- Doctoral Program in Morphological Sciences, Faculty of Medicine, Universidad de la Frontera, Temuco 4811230, Chile;
- Center of Molecular Biology and Pharmacogenetics, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4811230, Chile; (P.L.); (F.L.)
| | - Pía Loren
- Center of Molecular Biology and Pharmacogenetics, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4811230, Chile; (P.L.); (F.L.)
| | - Cristian Sandoval
- Escuela de Tecnología Médica, Facultad de Salud, Universidad Santo Tomás, Los Carreras 753, Osorno 5310431, Chile;
- Department of Internal Medicine, Faculty of Medicine, Universidad de La Frontera, Temuco 4811230, Chile
| | - Fernando Lanas
- Center of Molecular Biology and Pharmacogenetics, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4811230, Chile; (P.L.); (F.L.)
- Department of Internal Medicine, Faculty of Medicine, Universidad de La Frontera, Temuco 4811230, Chile
| | - Luis A. Salazar
- Center of Molecular Biology and Pharmacogenetics, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4811230, Chile; (P.L.); (F.L.)
| |
Collapse
|
300
|
Zhao HW, Jiang F, Chen S, Hu J, Xiang SH, Ding WY, Lu W, Tan B. Organocatalytic Asymmetric Construction and Application of Axially Chiral Spiro-bisindoles. Angew Chem Int Ed Engl 2025; 64:e202422951. [PMID: 39672796 DOI: 10.1002/anie.202422951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/11/2024] [Accepted: 12/13/2024] [Indexed: 12/15/2024]
Abstract
Spiro skeletons have emerged as a privileged class of chiral carriers across various research fields, including asymmetric catalysis and functional materials, due to their remarkable configurational rigidity. However, limited structural diversity of spiro frameworks significantly restricts the expansion of their applications. Here we present a new class of axially chiral spiro-bisindole frameworks and report their first enantioselective construction via a chiral phosphoric acid-catalyzed intramolecular dehydrative cyclization reaction. Unlike the classical SPINOL backbone, incorporation of indole moieties in place of phenol enhances the nucleophilicity of ketone substrates, thereby eliminating the need for a tedious pre-activation process. By leveraging the retained active sites of indole, the resulting highly enantioenriched spiro-bisindoles can be rapidly transformed into other valuable structures. More importantly, axially chiral fluorescent molecules with good asymmetry factors and quantum fluorescence efficiency are readily accessed, opening a new avenue for developing chiral fluorescent materials. Control experiments demonstrate the pivotal role of both unmasked N-H bonds in achieving good efficiency and enantiocontrol.
Collapse
Affiliation(s)
- Hao-Wen Zhao
- Shenzhen Grubbs Institute and Department of Chemistry, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Fei Jiang
- Shenzhen Grubbs Institute and Department of Chemistry, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Sihan Chen
- Shenzhen Grubbs Institute and Department of Chemistry, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Jingliang Hu
- Shenzhen Grubbs Institute and Department of Chemistry, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Shao-Hua Xiang
- Shenzhen Grubbs Institute and Department of Chemistry, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Wei-Yi Ding
- Shenzhen Grubbs Institute and Department of Chemistry, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Wei Lu
- Shenzhen Grubbs Institute and Department of Chemistry, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Bin Tan
- Shenzhen Grubbs Institute and Department of Chemistry, Southern University of Science and Technology, Shenzhen, 518055, China
| |
Collapse
|