Pub Date : 2025-12-04eCollection Date: 2025-01-01DOI: 10.3389/fcell.2025.1706393
Junyao Li, Lin Zhang, Yicun Wang, Chunyan Li
Circular RNAs (circRNAs), a class of non-coding RNAs characterized by covalently closed-loop structures, have emerged as key regulators in the tumor immune microenvironment (TIME) of lung cancer, owing to their high stability, tissue-specific expression, and multidimensional regulatory capabilities. This review systematically synthesizes the latest research progress and elucidates the processes by which circRNAs regulate the functional states of immune cells in the TIME through diverse molecular mechanisms, including acting as competing endogenous RNAs (ceRNAs) to sequester microRNAs (miRNAs), interacting with RNA-binding proteins (RBPs), and in some cases, encoding functional polypeptides. CircRNAs possess bidirectional regulatory capacities: they can promote tumor immune evasion by modulating the expression of immune checkpoint molecules, influencing the infiltration and activity of effector immune cells (e.g., CD8+ T cells), recruiting immunosuppressive cells (e.g., regulatory T cells and M2-type macrophages), and regulating immune signaling pathways; meanwhile, they can also activate antitumor immune responses. Furthermore, the review explores the potential of circRNAs as liquid biopsy biomarkers for lung cancer diagnosis and prognosis, as well as their translational prospects in therapeutic strategies including vaccines, circRNA-enhanced CAR-T therapy, and formulations encoding immunomodulatory factors. Despite challenges such as complex mechanisms, low delivery efficiency, and safety concerns, the development of multi-omics technologies, novel delivery systems, and gene-editing tools provides directions for the development of precision therapies targeting circRNAs, which aim to reshape the lung cancer immune microenvironment and overcome immunotherapy resistance.
{"title":"Dual-faced circRNAs: orchestrating immunosuppression and activation in the lung cancer microenvironment.","authors":"Junyao Li, Lin Zhang, Yicun Wang, Chunyan Li","doi":"10.3389/fcell.2025.1706393","DOIUrl":"10.3389/fcell.2025.1706393","url":null,"abstract":"<p><p>Circular RNAs (circRNAs), a class of non-coding RNAs characterized by covalently closed-loop structures, have emerged as key regulators in the tumor immune microenvironment (TIME) of lung cancer, owing to their high stability, tissue-specific expression, and multidimensional regulatory capabilities. This review systematically synthesizes the latest research progress and elucidates the processes by which circRNAs regulate the functional states of immune cells in the TIME through diverse molecular mechanisms, including acting as competing endogenous RNAs (ceRNAs) to sequester microRNAs (miRNAs), interacting with RNA-binding proteins (RBPs), and in some cases, encoding functional polypeptides. CircRNAs possess bidirectional regulatory capacities: they can promote tumor immune evasion by modulating the expression of immune checkpoint molecules, influencing the infiltration and activity of effector immune cells (e.g., CD8<sup>+</sup> T cells), recruiting immunosuppressive cells (e.g., regulatory T cells and M2-type macrophages), and regulating immune signaling pathways; meanwhile, they can also activate antitumor immune responses. Furthermore, the review explores the potential of circRNAs as liquid biopsy biomarkers for lung cancer diagnosis and prognosis, as well as their translational prospects in therapeutic strategies including vaccines, circRNA-enhanced CAR-T therapy, and formulations encoding immunomodulatory factors. Despite challenges such as complex mechanisms, low delivery efficiency, and safety concerns, the development of multi-omics technologies, novel delivery systems, and gene-editing tools provides directions for the development of precision therapies targeting circRNAs, which aim to reshape the lung cancer immune microenvironment and overcome immunotherapy resistance.</p>","PeriodicalId":12448,"journal":{"name":"Frontiers in Cell and Developmental Biology","volume":"13 ","pages":"1706393"},"PeriodicalIF":4.6,"publicationDate":"2025-12-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12713122/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145803913","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-04eCollection Date: 2025-01-01DOI: 10.3389/fcell.2025.1695775
Caiyu Liu, Xiaoying Xu, Hao Gu, Chi Wang, Lu Lu, Kaizhen Ye, Yan Zheng, Haiyan Wang, Wei Chen, Meiyan Li, Xiaolin Zhou, Shangkun Ou, Fangwen Yang
Objective: The aim of this study was to conduct a systematic investigation into the effects of ocular rotation on postoperative residual astigmatism in patients undergoing small incision lenticule extraction (SMILE).
Methods: A prospective observational cohort study involved 79 patients (153 eyes) with myopia and astigmatism who underwent SMILE surgery. Ocular rotational magnitude was measured using manual corneal and scleral marking with a slit-lamp microscopy assessment. Preoperative and postoperative (1- and 3-month) assessments included uncorrected distance visual acuity (UDVA), best-corrected visual acuity (BCVA), refractive error, and other relevant ocular parameters.
Results: Residual astigmatism showed significant correlations with ocular rotation magnitude (r = 0.429, p < 0.001), preoperative intraocular pressure (r = -0.178, p = 0.032), and preoperative cylindrical lens power (r = 0.175, p = 0.035). A multiple linear regression analysis indicated that rotation magnitude significantly impacted postoperative residual astigmatism (p < 0.001). However, preoperative intraocular pressure (p = 0.349) and spherical equivalent (p = 0.105) were not significantly related to residual astigmatism. Linear regression analysis further demonstrated significant positive correlations between rotation amplitude and various astigmatism parameters at both 1- and 3-month postoperative follow-ups (all p < 0.05). In particular, the relationships were quantified as follows: cylindrical lens (CYL [D]) (1 month: y = 7.058x + 17.480, p < 0.001; 3 months: y = 7.464x + 13.610, p < 0.001), target-induced astigmatism (TIA [D]) (1 month: y = 0.112x + 1.275, p = 0.012; 3 months: y = 0.097x + 1.217, p = 0.026), surgically induced astigmatism (SIA [D]) (1 month: y = 0.094x + 0.936, p < 0.001; 3 months: y = 0.059x + 0.911, p = 0.022), and difference vector (DV [D]) (1 month: y = 0.041x + 0.289, p = 0.005; 3 months: y = 0.037x + 0.866, p = 0.011). Notably, rotation amplitude exhibited the strongest association with postoperative CYL. Receiver operating characteristic (ROC) analysis determined the optimal thresholds for rotation magnitude in predicting residual astigmatism to be 1.5° at 1 month (AUC = 0.753; sensitivity 79.7%; specificity 58.2%) and 2.5° at 3 months (AUC = 0.929; sensitivity 92.9%; specificity 83.5%).
Conclusion: The magnitude of rotation shows a notably positive correlation with residual astigmatism during both the 1- and 3-month postoperative follow-ups. Thresholds of 1.5° (1 month) or 2.5° (3 months) prove predictive of residual astigmatism, with enhanced diagnostic precision at the later follow-up.
目的:本研究旨在系统探讨小切口晶状体摘除(SMILE)术后旋转对残余散光的影响。方法:对79例(153眼)近视散光行SMILE手术的患者进行前瞻性观察队列研究。通过手工角膜和巩膜标记和裂隙灯显微镜评估来测量眼球旋转幅度。术前和术后(1个月和3个月)评估包括未矫正距离视力(UDVA)、最佳矫正视力(BCVA)、屈光不正和其他相关眼部参数。结果:剩余散光与眼旋转幅度(r = 0.429, p < 0.001)、术前眼压(r = -0.178, p = 0.032)、术前柱状晶状体度数(r = 0.175, p = 0.035)有显著相关性。多元线性回归分析显示旋转程度显著影响术后残留散光(p < 0.001)。术前眼压(p = 0.349)和球当量(p = 0.105)与剩余散光无显著相关。线性回归分析进一步表明,术后1个月和3个月随访时旋转幅度与各项散光参数呈正相关(均p < 0.05)。特别是,被量化的关系如下:柱面透镜(共青团[D])(1个月:y = 7.058 x + 17.480, p < 0.001; 3个月:y = 7.464 x + 13.610, p < 0.001), target-induced散光(TIA [D])(1个月:y = 0.112 x + 1.275, p = 0.012; 3个月:y = 0.097 x + 1.217, p = 0.026),手术引起散光(新航[D])(1个月:y = 0.094 x + 0.936, p < 0.001; 3个月:y = 0.059 x + 0.911, p = 0.022),和差分向量(DV [D])(1个月:y = 0.041 x + 0.289, p = 0.005;3个月:y = 0.037x + 0.866, p = 0.011)。值得注意的是,旋转幅度与术后CYL的相关性最强。受试者工作特征(ROC)分析确定,旋转幅度预测残留散光的最佳阈值为1个月时1.5°(AUC = 0.753,灵敏度79.7%,特异性58.2%)和3个月时2.5°(AUC = 0.929,灵敏度92.9%,特异性83.5%)。结论:在术后1个月和3个月的随访中,旋转大小与剩余散光有显著正相关。1.5°(1个月)或2.5°(3个月)的阈值可预测残余散光,在后期随访时诊断精度提高。
{"title":"Quantitative analysis of the effect of ocular rotation on postoperative residual astigmatism in small incision lenticule extraction for myopia correction.","authors":"Caiyu Liu, Xiaoying Xu, Hao Gu, Chi Wang, Lu Lu, Kaizhen Ye, Yan Zheng, Haiyan Wang, Wei Chen, Meiyan Li, Xiaolin Zhou, Shangkun Ou, Fangwen Yang","doi":"10.3389/fcell.2025.1695775","DOIUrl":"10.3389/fcell.2025.1695775","url":null,"abstract":"<p><strong>Objective: </strong>The aim of this study was to conduct a systematic investigation into the effects of ocular rotation on postoperative residual astigmatism in patients undergoing small incision lenticule extraction (SMILE).</p><p><strong>Methods: </strong>A prospective observational cohort study involved 79 patients (153 eyes) with myopia and astigmatism who underwent SMILE surgery. Ocular rotational magnitude was measured using manual corneal and scleral marking with a slit-lamp microscopy assessment. Preoperative and postoperative (1- and 3-month) assessments included uncorrected distance visual acuity (UDVA), best-corrected visual acuity (BCVA), refractive error, and other relevant ocular parameters.</p><p><strong>Results: </strong>Residual astigmatism showed significant correlations with ocular rotation magnitude (r = 0.429, <i>p</i> < 0.001), preoperative intraocular pressure (r = -0.178, <i>p</i> = 0.032), and preoperative cylindrical lens power (r = 0.175, <i>p</i> = 0.035). A multiple linear regression analysis indicated that rotation magnitude significantly impacted postoperative residual astigmatism (<i>p</i> < 0.001). However, preoperative intraocular pressure (<i>p</i> = 0.349) and spherical equivalent (<i>p</i> = 0.105) were not significantly related to residual astigmatism. Linear regression analysis further demonstrated significant positive correlations between rotation amplitude and various astigmatism parameters at both 1- and 3-month postoperative follow-ups (all <i>p</i> < 0.05). In particular, the relationships were quantified as follows: cylindrical lens (CYL [D]) (1 month: y = 7.058x + 17.480, <i>p</i> < 0.001; 3 months: y = 7.464x + 13.610, <i>p</i> < 0.001), target-induced astigmatism (TIA [D]) (1 month: y = 0.112x + 1.275, <i>p</i> = 0.012; 3 months: y = 0.097x + 1.217, <i>p</i> = 0.026), surgically induced astigmatism (SIA [D]) (1 month: y = 0.094x + 0.936, <i>p</i> < 0.001; 3 months: y = 0.059x + 0.911, <i>p</i> = 0.022), and difference vector (DV [D]) (1 month: y = 0.041x + 0.289, <i>p</i> = 0.005; 3 months: y = 0.037x + 0.866, <i>p</i> = 0.011). Notably, rotation amplitude exhibited the strongest association with postoperative CYL. Receiver operating characteristic (ROC) analysis determined the optimal thresholds for rotation magnitude in predicting residual astigmatism to be 1.5° at 1 month (AUC = 0.753; sensitivity 79.7%; specificity 58.2%) and 2.5° at 3 months (AUC = 0.929; sensitivity 92.9%; specificity 83.5%).</p><p><strong>Conclusion: </strong>The magnitude of rotation shows a notably positive correlation with residual astigmatism during both the 1- and 3-month postoperative follow-ups. Thresholds of 1.5° (1 month) or 2.5° (3 months) prove predictive of residual astigmatism, with enhanced diagnostic precision at the later follow-up.</p>","PeriodicalId":12448,"journal":{"name":"Frontiers in Cell and Developmental Biology","volume":"13 ","pages":"1695775"},"PeriodicalIF":4.6,"publicationDate":"2025-12-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12713198/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145803892","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-04eCollection Date: 2025-01-01DOI: 10.3389/fcell.2025.1719978
Anna Di Spirito, Gaia Zuccolotto, Anna Tosi, Sahar Balkhi, Antonio Rosato, Denisa Baci, Lorenzo Mortara
Bladder cancer (BC) remains a prevalent malignancy with high recurrence rates despite standard therapies. Bacille Calmette-Guérin (BCG) is the cornerstone of treatment for non-muscle-invasive bladder cancer (NMIBC); however, nearly half of patients experience relapse or develop resistance, highlighting the need for alternative strategies. Recent advances in immunotherapy have reshaped the therapeutic landscape. Immune checkpoint inhibitors (ICIs) restore T-cell function and show clinical activity in BCG-unresponsive disease. Viral vector-based approaches, including nadofaragene firadenovec and CG0070, provide localized immune activation, while cellular platforms such as CAR-T and CAR-NK therapies offer precision targeting of tumor antigens. Concurrently, nanotechnology-based delivery systems and antibody-drug conjugates (ADCs) enhance efficacy and safety by improving tumor-specific cytotoxicity. Collectively, these strategies signify a paradigm shift from traditional intravesical therapy toward personalized and durable immunotherapeutic interventions. Identification of predictive biomarkers and rational combination strategies will be critical to improving outcomes and guiding the future management of BC.
{"title":"Evolving frontiers in bladder cancer immunotherapy: integrating BCG, immune checkpoints, viral vectors, nanotechnology, and CAR-based therapies.","authors":"Anna Di Spirito, Gaia Zuccolotto, Anna Tosi, Sahar Balkhi, Antonio Rosato, Denisa Baci, Lorenzo Mortara","doi":"10.3389/fcell.2025.1719978","DOIUrl":"10.3389/fcell.2025.1719978","url":null,"abstract":"<p><p>Bladder cancer (BC) remains a prevalent malignancy with high recurrence rates despite standard therapies. Bacille Calmette-Guérin (BCG) is the cornerstone of treatment for non-muscle-invasive bladder cancer (NMIBC); however, nearly half of patients experience relapse or develop resistance, highlighting the need for alternative strategies. Recent advances in immunotherapy have reshaped the therapeutic landscape. Immune checkpoint inhibitors (ICIs) restore T-cell function and show clinical activity in BCG-unresponsive disease. Viral vector-based approaches, including nadofaragene firadenovec and CG0070, provide localized immune activation, while cellular platforms such as CAR-T and CAR-NK therapies offer precision targeting of tumor antigens. Concurrently, nanotechnology-based delivery systems and antibody-drug conjugates (ADCs) enhance efficacy and safety by improving tumor-specific cytotoxicity. Collectively, these strategies signify a paradigm shift from traditional intravesical therapy toward personalized and durable immunotherapeutic interventions. Identification of predictive biomarkers and rational combination strategies will be critical to improving outcomes and guiding the future management of BC.</p>","PeriodicalId":12448,"journal":{"name":"Frontiers in Cell and Developmental Biology","volume":"13 ","pages":"1719978"},"PeriodicalIF":4.6,"publicationDate":"2025-12-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12711830/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145803910","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Proliferative vitreoretinopathy (PVR) is a major complication of rhegmatogenous retinal detachment. Epithelial-mesenchymal transition (EMT) of retinal pigment epithelial (RPE) plays a central role in PVR pathogenesis. This study aims to investigate the effect of ADP-ribosylation factor-like 13B (ARL13B) on RPE EMT in PVR.
Methods: The expression of ARL13B in PVR specimens was analyzed by immunofluorescence (IF) staining. The effect of ARL13B on RPE EMT was assessed by IF staining and Western blot. The proliferation and migration of RPE were measured with EdU and transwell and scratch assays, respectively. The EMT-related transcriptome was analyzed by bulk RNAseq. An intravitreal injection mouse model of PVR was used to investigate the role of ARL13B in PVR formation.
Results: Immunofluorescence revealed significantly reduced ARL13B levels in α-SMA-positive cells as compared with Pan-CK-positive cells in an epiretinal membrane derived from retinal tears. During EMT, TGFβ1 treatment remarkably reduced ARL13B expression and shortened the length of cilia in RPE cells. In line with this, ARL13B knockdown (KD) decreased the length of cilia and enhanced TGFβ1-induced EMT, evidenced by morphology change and a globally upregulated EMT-related gene expression in RPEs. Moreover, ARL13B KD enhanced TGFβ1-induced RPE proliferation and migration. Consistently, ARL13B KD promoted PVR formation in vivo. Mechanistically, ARL13B KD enhanced TGFβ1 signaling by increasing the phosphorylation and expression of Smad3.
Conclusion: This study demonstrated a crucial role of ARL13B on TGFβ1-induced RPE EMT, highlighting the importance of ARL13B in PVR formation.
{"title":"Loss of a primary cilia protein ARL13B promotes TGFβ-1 induced EMT of RPE in proliferative vitreoretinopathy via increasing Smad3 expression.","authors":"Wenjun Sui, Xinqi Ma, Yajun Gong, Kairui Qiu, Miner Yuan, Keli Mao, Xiaobing Qian, Jieting Zeng, Yujie Li, Bingsheng Lou, Xiaofeng Lin, Xiaolai Zhou","doi":"10.3389/fcell.2025.1661658","DOIUrl":"10.3389/fcell.2025.1661658","url":null,"abstract":"<p><strong>Background: </strong>Proliferative vitreoretinopathy (PVR) is a major complication of rhegmatogenous retinal detachment. Epithelial-mesenchymal transition (EMT) of retinal pigment epithelial (RPE) plays a central role in PVR pathogenesis. This study aims to investigate the effect of ADP-ribosylation factor-like 13B (ARL13B) on RPE EMT in PVR.</p><p><strong>Methods: </strong>The expression of ARL13B in PVR specimens was analyzed by immunofluorescence (IF) staining. The effect of ARL13B on RPE EMT was assessed by IF staining and Western blot. The proliferation and migration of RPE were measured with EdU and transwell and scratch assays, respectively. The EMT-related transcriptome was analyzed by bulk RNAseq. An intravitreal injection mouse model of PVR was used to investigate the role of ARL13B in PVR formation.</p><p><strong>Results: </strong>Immunofluorescence revealed significantly reduced ARL13B levels in α-SMA-positive cells as compared with Pan-CK-positive cells in an epiretinal membrane derived from retinal tears. During EMT, TGFβ1 treatment remarkably reduced ARL13B expression and shortened the length of cilia in RPE cells. In line with this, ARL13B knockdown (KD) decreased the length of cilia and enhanced TGFβ1-induced EMT, evidenced by morphology change and a globally upregulated EMT-related gene expression in RPEs. Moreover, ARL13B KD enhanced TGFβ1-induced RPE proliferation and migration. Consistently, ARL13B KD promoted PVR formation <i>in vivo</i>. Mechanistically, ARL13B KD enhanced TGFβ1 signaling by increasing the phosphorylation and expression of Smad3.</p><p><strong>Conclusion: </strong>This study demonstrated a crucial role of ARL13B on TGFβ1-induced RPE EMT, highlighting the importance of ARL13B in PVR formation.</p>","PeriodicalId":12448,"journal":{"name":"Frontiers in Cell and Developmental Biology","volume":"13 ","pages":"1661658"},"PeriodicalIF":4.6,"publicationDate":"2025-12-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12711727/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145803873","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-03eCollection Date: 2025-01-01DOI: 10.3389/fcell.2025.1698265
Qian Zhou, Minxin He, Min Liu, Guoying Sun, Jian Li
tRNA-derived small RNAs (tsRNAs) are a class of non-coding RNAs(ncRNAs) generated from precursor or mature tRNAs under stress conditions, such as starvation, hypoxia, or oxidative stress. They are broadly classified into a growing class of small RNAs, known as tRNA-derived RNA (tDR), tRNA-derived small RNAs or tRNA-derived fragments and tRNA-derived stress-induced RNAs (tiRNAs) based on their cleavage sites. Recent advances in high-throughput sequencing have revealed their critical roles in the reproductive system, particularly in spermatogenesis, sperm maturation, and male infertility. In females, tsRNAs are implicated in oocyte development and embryo implantation. Dysregulation of tsRNAs has also been linked to reproductive diseases, including polycystic ovary syndrome (PCOS) and endometriosis in women, and oligospermia or azoospermia in men. Mechanistically, tsRNAs regulate gene expression, mRNA stability, and translation, influencing key pathways in reproductive health and disease. Their potential as biomarkers and therapeutic targets for reproductive disorders is increasingly recognized, though further research is needed to fully elucidate their roles and clinical applications.
{"title":"Emerging roles and therapeutic potential of tRNA-Derived small RNAs in reproductive system diseases: a review.","authors":"Qian Zhou, Minxin He, Min Liu, Guoying Sun, Jian Li","doi":"10.3389/fcell.2025.1698265","DOIUrl":"10.3389/fcell.2025.1698265","url":null,"abstract":"<p><p>tRNA-derived small RNAs (tsRNAs) are a class of non-coding RNAs(ncRNAs) generated from precursor or mature tRNAs under stress conditions, such as starvation, hypoxia, or oxidative stress. They are broadly classified into a growing class of small RNAs, known as tRNA-derived RNA (tDR), tRNA-derived small RNAs or tRNA-derived fragments and tRNA-derived stress-induced RNAs (tiRNAs) based on their cleavage sites. Recent advances in high-throughput sequencing have revealed their critical roles in the reproductive system, particularly in spermatogenesis, sperm maturation, and male infertility. In females, tsRNAs are implicated in oocyte development and embryo implantation. Dysregulation of tsRNAs has also been linked to reproductive diseases, including polycystic ovary syndrome (PCOS) and endometriosis in women, and oligospermia or azoospermia in men. Mechanistically, tsRNAs regulate gene expression, mRNA stability, and translation, influencing key pathways in reproductive health and disease. Their potential as biomarkers and therapeutic targets for reproductive disorders is increasingly recognized, though further research is needed to fully elucidate their roles and clinical applications.</p>","PeriodicalId":12448,"journal":{"name":"Frontiers in Cell and Developmental Biology","volume":"13 ","pages":"1698265"},"PeriodicalIF":4.6,"publicationDate":"2025-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12708580/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145780734","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Introduction: Tumor necrosis factor-α (TNF-α) is considered a potential therapeutic strategy for cancers, as it exacerbates calcium influx through voltage-gated calcium channels (VGCCs), thereby inducing apoptosis. However, the mechanisms underlying TNF-α's effects at the single-molecule level remain unclear.
Methods: This study employed multiple modes of Atomic Force Microscopy (AFM) to investigate the impact of TNF-α on breast cancer cells. The measurements were performed with nanometer spatial resolution, picoNewton force sensitivity, picoAmpere current precision, and 0.1 mV surface potential accuracy.
Results: The results revealed that TNF-α treatment significantly increased the density and aggregation of VGCCs on the cell membrane while enhancing their channel activity. Concurrently, the electrical conductivity and surface potential of the membrane were elevated, collectively promoting exacerbated calcium influx.
Discussion: These findings elucidate the mechanisms by which TNF-α modulates VGCC distribution and electrophysiological properties to amplify calcium signaling, ultimately triggering apoptosis. This study provides unprecedented insights into TNF-α-induced calcium dysregulation in cancer cells at the single-molecule level, offering a novel approach for investigating apoptosis and advancing targeted therapies for breast cancer and other malignancies.
{"title":"TNF-α exacerbates calcium influx via voltage-gated calcium channels in breast cancer cells: a nanoscale multimodal AFM study.","authors":"Zhongwei Wang, Qianhui Xu, Huaiwei Zhang, Rongrong Feng, Junmei Chen, Jinsong Wei, Haijian Zhong, Weidong Zhao","doi":"10.3389/fcell.2025.1633976","DOIUrl":"10.3389/fcell.2025.1633976","url":null,"abstract":"<p><strong>Introduction: </strong>Tumor necrosis factor-α (TNF-α) is considered a potential therapeutic strategy for cancers, as it exacerbates calcium influx through voltage-gated calcium channels (VGCCs), thereby inducing apoptosis. However, the mechanisms underlying TNF-α's effects at the single-molecule level remain unclear.</p><p><strong>Methods: </strong>This study employed multiple modes of Atomic Force Microscopy (AFM) to investigate the impact of TNF-α on breast cancer cells. The measurements were performed with nanometer spatial resolution, picoNewton force sensitivity, picoAmpere current precision, and 0.1 mV surface potential accuracy.</p><p><strong>Results: </strong>The results revealed that TNF-α treatment significantly increased the density and aggregation of VGCCs on the cell membrane while enhancing their channel activity. Concurrently, the electrical conductivity and surface potential of the membrane were elevated, collectively promoting exacerbated calcium influx.</p><p><strong>Discussion: </strong>These findings elucidate the mechanisms by which TNF-α modulates VGCC distribution and electrophysiological properties to amplify calcium signaling, ultimately triggering apoptosis. This study provides unprecedented insights into TNF-α-induced calcium dysregulation in cancer cells at the single-molecule level, offering a novel approach for investigating apoptosis and advancing targeted therapies for breast cancer and other malignancies.</p>","PeriodicalId":12448,"journal":{"name":"Frontiers in Cell and Developmental Biology","volume":"13 ","pages":"1633976"},"PeriodicalIF":4.6,"publicationDate":"2025-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12708539/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145780647","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Fatty acids are not only important as energy sources, but also playing crucial roles in maintaining cellular homeostasis. However, their role in preimplantation development remains unclear. Here, we find that fatty acids gradually accumulate after 4-cell stage in mouse preimplantation development. And, the expressions of fatty acid degradation-related genes are increased along the developmental process. Inhibition of long-chain fatty acid β-oxidation (LCFAO) results in preimplantation developmental arrest, downregulated expressions of S phase-related genes, and loss of H3K18ac modification. By profiling the landscape of H3K18ac, we show that H3K18ac is enriched on the promoter regions of S phase-related genes and correlates with their expression. Together, these findings suggest that LCFAO regulate mouse preimplantation development through H3K18ac, providing additional evidence for metabolic-epigenetic crosstalk in embryonic development.
{"title":"Long-chain fatty acid β-oxidation regulates embryonic development by H3K18 acetylation in mice.","authors":"Kefan Zheng, Hongdi Cui, Ziheng Tang, Entong Song, Qingran Kong, Jiaming Zhang, Hao Li, Qi Zhao","doi":"10.3389/fcell.2025.1683028","DOIUrl":"10.3389/fcell.2025.1683028","url":null,"abstract":"<p><p>Fatty acids are not only important as energy sources, but also playing crucial roles in maintaining cellular homeostasis. However, their role in preimplantation development remains unclear. Here, we find that fatty acids gradually accumulate after 4-cell stage in mouse preimplantation development. And, the expressions of fatty acid degradation-related genes are increased along the developmental process. Inhibition of long-chain fatty acid β-oxidation (LCFAO) results in preimplantation developmental arrest, downregulated expressions of S phase-related genes, and loss of H3K18ac modification. By profiling the landscape of H3K18ac, we show that H3K18ac is enriched on the promoter regions of S phase-related genes and correlates with their expression. Together, these findings suggest that LCFAO regulate mouse preimplantation development through H3K18ac, providing additional evidence for metabolic-epigenetic crosstalk in embryonic development.</p>","PeriodicalId":12448,"journal":{"name":"Frontiers in Cell and Developmental Biology","volume":"13 ","pages":"1683028"},"PeriodicalIF":4.6,"publicationDate":"2025-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12708569/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145780706","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-03eCollection Date: 2025-01-01DOI: 10.3389/fcell.2025.1647255
Tatyana F Kovalenko, Amal Abdurazakov, Nadezhda V Antipova, Mikhail I Shakhparonov, Marat S Pavlyukov
R-loops consist of double-stranded DNA-RNA hybrids and a complementary DNA strand that is displaced from the duplex. R-loops play important role in numerous normal physiological processes, including DNA methylation, chromatin remodeling, RNA editing, replication, DNA repair, immunoglobulin class switching, and chromosome segregation during cell division. However, excessive or untimely formation of R-loops can lead to replicative collapse and subsequent DNA damage, resulting in genomic instability. One type of genomic rearrangements that is strongly associated with cancer malignancy is the extrachromosomal amplification of genes on circular DNA molecules (ecDNA). These molecules are relieved of hereditary constraints and conventional segregation laws and can endow cancer cells with the ability to rapidly change their genome, thereby accelerating tumor evolution and the development of therapy resistance. Multiple lines of evidence indicate that upregulated transcription of a gene can increase its susceptibility to amplification. Although the mechanisms underlying these processes are not yet fully understood, R-loops may play an important role in initiating gene amplification. In this review, we highlight the role of R-loops in replicative collapse, double-strand breaks, and DNA damage repair. We also provide examples of gene amplifications that is known to be induced by R-loops. Finally, we discuss amplification mechanisms in which involvement of R-loops has not yet been demonstrated, but appears highly likely.
{"title":"R-loops as a trigger for intra- and extrachromosomal DNA amplification in cancer.","authors":"Tatyana F Kovalenko, Amal Abdurazakov, Nadezhda V Antipova, Mikhail I Shakhparonov, Marat S Pavlyukov","doi":"10.3389/fcell.2025.1647255","DOIUrl":"10.3389/fcell.2025.1647255","url":null,"abstract":"<p><p>R-loops consist of double-stranded DNA-RNA hybrids and a complementary DNA strand that is displaced from the duplex. R-loops play important role in numerous normal physiological processes, including DNA methylation, chromatin remodeling, RNA editing, replication, DNA repair, immunoglobulin class switching, and chromosome segregation during cell division. However, excessive or untimely formation of R-loops can lead to replicative collapse and subsequent DNA damage, resulting in genomic instability. One type of genomic rearrangements that is strongly associated with cancer malignancy is the extrachromosomal amplification of genes on circular DNA molecules (ecDNA). These molecules are relieved of hereditary constraints and conventional segregation laws and can endow cancer cells with the ability to rapidly change their genome, thereby accelerating tumor evolution and the development of therapy resistance. Multiple lines of evidence indicate that upregulated transcription of a gene can increase its susceptibility to amplification. Although the mechanisms underlying these processes are not yet fully understood, R-loops may play an important role in initiating gene amplification. In this review, we highlight the role of R-loops in replicative collapse, double-strand breaks, and DNA damage repair. We also provide examples of gene amplifications that is known to be induced by R-loops. Finally, we discuss amplification mechanisms in which involvement of R-loops has not yet been demonstrated, but appears highly likely.</p>","PeriodicalId":12448,"journal":{"name":"Frontiers in Cell and Developmental Biology","volume":"13 ","pages":"1647255"},"PeriodicalIF":4.6,"publicationDate":"2025-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12708599/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145780718","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-03eCollection Date: 2025-01-01DOI: 10.3389/fcell.2025.1718799
Yiming Liu, Yue Wang, Jiaying Zhou, Hong Li, Caiyun Liu, Beilei Zhong, Juan Liu, Leiming Liu, Lingling Zhang, Leimin Sun
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a complex and heterogeneous metabolic disorder where subcellular organelle dysfunction and disrupted inter-organelle communication are recognized as increasingly important drivers of pathogenesis, moving beyond traditional views focused solely on macroscopic metabolic regulation. This review systematically explores the functional impairments of key organelles-including mitochondria, the endoplasmic reticulum, lipid droplets, and autophagic pathways-to delineate their collective roles in fostering lipid metabolism imbalance, oxidative stress, and inflammation. A key innovation discussed is how the pathological dysregulation of membrane contact sites (MCSs) acts as a pivotal mechanism decoupling organelle function and accelerating disease progression. We conclude that therapeutic strategies aimed at restoring cellular metabolic flexibility-by precisely modulating MCSs, activating clearance pathways, and restoring energy metabolism-represent a promising new paradigm for treating MASLD, particularly in patient populations unresponsive to current therapies.
{"title":"Restoring metabolic flexibility: targeting organelle interaction networks in the pathogenesis and therapy of MASLD.","authors":"Yiming Liu, Yue Wang, Jiaying Zhou, Hong Li, Caiyun Liu, Beilei Zhong, Juan Liu, Leiming Liu, Lingling Zhang, Leimin Sun","doi":"10.3389/fcell.2025.1718799","DOIUrl":"10.3389/fcell.2025.1718799","url":null,"abstract":"<p><p>Metabolic dysfunction-associated steatotic liver disease (MASLD) is a complex and heterogeneous metabolic disorder where subcellular organelle dysfunction and disrupted inter-organelle communication are recognized as increasingly important drivers of pathogenesis, moving beyond traditional views focused solely on macroscopic metabolic regulation. This review systematically explores the functional impairments of key organelles-including mitochondria, the endoplasmic reticulum, lipid droplets, and autophagic pathways-to delineate their collective roles in fostering lipid metabolism imbalance, oxidative stress, and inflammation. A key innovation discussed is how the pathological dysregulation of membrane contact sites (MCSs) acts as a pivotal mechanism decoupling organelle function and accelerating disease progression. We conclude that therapeutic strategies aimed at restoring cellular metabolic flexibility-by precisely modulating MCSs, activating clearance pathways, and restoring energy metabolism-represent a promising new paradigm for treating MASLD, particularly in patient populations unresponsive to current therapies.</p>","PeriodicalId":12448,"journal":{"name":"Frontiers in Cell and Developmental Biology","volume":"13 ","pages":"1718799"},"PeriodicalIF":4.6,"publicationDate":"2025-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12708529/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145780727","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-03eCollection Date: 2025-01-01DOI: 10.3389/fcell.2025.1712195
Feng Yu, Bangwei Che, Wei Li
<p><strong>Background: </strong>In recent years, growing attention has been paid to the carcinogenicity of endocrine disruptors (EDs). However, their relationship with prostate cancer (PCa) remains unclear. This study investigates the association between EDs and PCa to identify key genes that may bridge this relationship.</p><p><strong>Methods: </strong>The ADME properties and carcinogenicity of the selected endocrine-disrupting chemicals EDs were predicted using the ADMETlab 3.0 and ProTox 3.0 platforms, respectively. Potential target genes related to EDs and PCa were obtained by integrating multiple public databases. A protein-protein interaction (PPI) network of the overlapping genes was constructed and visualized, followed by GO and KEGG enrichment analyses to explore their potential biological mechanisms. From 101 machine learning algorithm combinations, the most relevant key genes for PCa progression were screened. Molecular docking analysis was used to evaluate the binding properties between ED compounds and key targets. Pan-cancer analysis was employed to examine the general role of key genes across multiple cancer types. The Comparative Toxicogenomics Database (CTD) was used to identify natural active products potentially targeting the core genes. Finally, <i>in vitro</i> cell experiments were conducted to validate the effects of EDs on PCa cells and the intervention effects of related natural products.</p><p><strong>Results: </strong>Initially, predictions from the ADMETlab 3.0 and ProTox 3.0 platforms indicated significant <i>in vivo</i> accumulation, endocrine-disrupting effects, and carcinogenicity for the 12 common EDs. Subsequently, the integration of multiple databases identified 233 overlapping targets associated with PCa. GO and KEGG enrichment analyses revealed that these targets are primarily involved in regulating cell proliferation, inflammatory responses, and cancer cell metabolism. Among the evaluated machine learning algorithms, the CoxBoost + SuperPC hybrid model demonstrated superior predictive performance and robustness. Subsequent analysis pinpointed three key regulatory genes: CD38, MMP11, and PLK1. Molecular docking simulations confirmed potential interactions between EDs compounds and the core target, PLK1. Furthermore, five natural active products were identified as potential agents to mitigate the adverse effects induced by EDs exposure. Finally, <i>in vitro</i> cell experiments demonstrated that Benzo[a]pyrene promotes PLK1 expression and PCa progression, whereas Cryptotanshinone effectively counteracts these effects.</p><p><strong>Conclusion: </strong>This multidisciplinary study unveils PLK1 as a pivotal molecular target through which EDs drive PCa progression. Furthermore, we identify five natural compounds, notably Cryptotanshinone, that counteract the carcinogenic effects of EDs by targeting PLK1. These findings provide crucial molecular insights into ED-induced carcinogenesis and reveal promising targets f
{"title":"Comprehensive bioinformatics and in vitro studies reveal the carcinogenic role and molecular basis of endocrine disruptors in prostate cancer.","authors":"Feng Yu, Bangwei Che, Wei Li","doi":"10.3389/fcell.2025.1712195","DOIUrl":"10.3389/fcell.2025.1712195","url":null,"abstract":"<p><strong>Background: </strong>In recent years, growing attention has been paid to the carcinogenicity of endocrine disruptors (EDs). However, their relationship with prostate cancer (PCa) remains unclear. This study investigates the association between EDs and PCa to identify key genes that may bridge this relationship.</p><p><strong>Methods: </strong>The ADME properties and carcinogenicity of the selected endocrine-disrupting chemicals EDs were predicted using the ADMETlab 3.0 and ProTox 3.0 platforms, respectively. Potential target genes related to EDs and PCa were obtained by integrating multiple public databases. A protein-protein interaction (PPI) network of the overlapping genes was constructed and visualized, followed by GO and KEGG enrichment analyses to explore their potential biological mechanisms. From 101 machine learning algorithm combinations, the most relevant key genes for PCa progression were screened. Molecular docking analysis was used to evaluate the binding properties between ED compounds and key targets. Pan-cancer analysis was employed to examine the general role of key genes across multiple cancer types. The Comparative Toxicogenomics Database (CTD) was used to identify natural active products potentially targeting the core genes. Finally, <i>in vitro</i> cell experiments were conducted to validate the effects of EDs on PCa cells and the intervention effects of related natural products.</p><p><strong>Results: </strong>Initially, predictions from the ADMETlab 3.0 and ProTox 3.0 platforms indicated significant <i>in vivo</i> accumulation, endocrine-disrupting effects, and carcinogenicity for the 12 common EDs. Subsequently, the integration of multiple databases identified 233 overlapping targets associated with PCa. GO and KEGG enrichment analyses revealed that these targets are primarily involved in regulating cell proliferation, inflammatory responses, and cancer cell metabolism. Among the evaluated machine learning algorithms, the CoxBoost + SuperPC hybrid model demonstrated superior predictive performance and robustness. Subsequent analysis pinpointed three key regulatory genes: CD38, MMP11, and PLK1. Molecular docking simulations confirmed potential interactions between EDs compounds and the core target, PLK1. Furthermore, five natural active products were identified as potential agents to mitigate the adverse effects induced by EDs exposure. Finally, <i>in vitro</i> cell experiments demonstrated that Benzo[a]pyrene promotes PLK1 expression and PCa progression, whereas Cryptotanshinone effectively counteracts these effects.</p><p><strong>Conclusion: </strong>This multidisciplinary study unveils PLK1 as a pivotal molecular target through which EDs drive PCa progression. Furthermore, we identify five natural compounds, notably Cryptotanshinone, that counteract the carcinogenic effects of EDs by targeting PLK1. These findings provide crucial molecular insights into ED-induced carcinogenesis and reveal promising targets f","PeriodicalId":12448,"journal":{"name":"Frontiers in Cell and Developmental Biology","volume":"13 ","pages":"1712195"},"PeriodicalIF":4.6,"publicationDate":"2025-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12708592/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145780694","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}