Pub Date : 2025-11-19eCollection Date: 2025-12-01DOI: 10.1016/j.bbrep.2025.102356
Thomas H Hampton, Lily Taub, Kiyoshi Ferreria-Fukutani, Bruce A Stanton, Todd A MacKenzie
Quantitative PCR (qPCR) remains a widely used, cost-effective method for RNA quantitation, yet many published studies inadequately comply with MIQE (Minimum Information for Publication of Quantitative Real-Time PCR Experiments) and FAIR (Findable, Accessible, Interoperable, Reproducible) data principles. Widespread reliance on the 2-ΔΔCT method often overlooks critical factors such as amplification efficiency variability and reference gene stability. Furthermore, the absence of raw data and analysis code limits the community's ability to evaluate potential biases and reproduce findings. Here, we primarily aim to encourage researchers to share raw qPCR fluorescence data along with detailed analysis scripts that start from raw input and produce final figures and statistical tests. Using our recently published dataset, we model the complete qPCR analytical workflow-from raw fluorescence curves through to differential expression-highlighting key decision points that can influence results. We provide fully documented R scripts illustrating how ANCOVA (Analysis of Covariance), a flexible multivariable linear modeling approach, generally offers greater statistical power and robustness compared to 2-ΔΔCT. Additionally, simulations support ANCOVA's applicability across diverse experimental conditions. We also demonstrate how general-purpose data repositories (e.g., figshare) and code repositories (e.g., GitHub) facilitate adherence to FAIR principles and promote transparency in qPCR research. Finally, we offer graphical examples that transparently depict both target and reference gene behavior within the same figure, enhancing interpretability. This work establishes practical resources and conceptual foundations to improve rigor, reproducibility, and openness in qPCR data analysis.Image 1.
{"title":"Analyzing qPCR data: Better practices to facilitate rigor and reproducibility.","authors":"Thomas H Hampton, Lily Taub, Kiyoshi Ferreria-Fukutani, Bruce A Stanton, Todd A MacKenzie","doi":"10.1016/j.bbrep.2025.102356","DOIUrl":"10.1016/j.bbrep.2025.102356","url":null,"abstract":"<p><p>Quantitative PCR (qPCR) remains a widely used, cost-effective method for RNA quantitation, yet many published studies inadequately comply with MIQE (Minimum Information for Publication of Quantitative Real-Time PCR Experiments) and FAIR (Findable, Accessible, Interoperable, Reproducible) data principles. Widespread reliance on the 2<sup>-ΔΔCT</sup> method often overlooks critical factors such as amplification efficiency variability and reference gene stability. Furthermore, the absence of raw data and analysis code limits the community's ability to evaluate potential biases and reproduce findings. Here, we primarily aim to encourage researchers to share raw qPCR fluorescence data along with detailed analysis scripts that start from raw input and produce final figures and statistical tests. Using our recently published dataset, we model the complete qPCR analytical workflow-from raw fluorescence curves through to differential expression-highlighting key decision points that can influence results. We provide fully documented R scripts illustrating how ANCOVA (Analysis of Covariance), a flexible multivariable linear modeling approach, generally offers greater statistical power and robustness compared to 2<sup>-ΔΔCT</sup>. Additionally, simulations support ANCOVA's applicability across diverse experimental conditions. We also demonstrate how general-purpose data repositories (e.g., figshare) and code repositories (e.g., GitHub) facilitate adherence to FAIR principles and promote transparency in qPCR research. Finally, we offer graphical examples that transparently depict both target and reference gene behavior within the same figure, enhancing interpretability. This work establishes practical resources and conceptual foundations to improve rigor, reproducibility, and openness in qPCR data analysis.Image 1.</p>","PeriodicalId":8771,"journal":{"name":"Biochemistry and Biophysics Reports","volume":"44 ","pages":"102356"},"PeriodicalIF":2.2,"publicationDate":"2025-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12666847/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145660082","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-19eCollection Date: 2025-12-01DOI: 10.1016/j.bbrep.2025.102352
Yijia He, Shuanghu Dong, Lifen Ouyang, Jun Kang, Liying Chen, Qingyun He
Sepsis, a pressing global health challenge, is characterized by immune dysregulation and high mortality. This study, in response to this urgent need, integrated Single-cell RNA sequencing data from 53 samples and bulk transcriptomic data from 479 sepsis patients to analyze immune microenvironment heterogeneity and develop a prognostic signature. Using the scVI algorithm, eight immune cell types were identified. B, T, and NK cells were reduced in sepsis, while neutrophils were elevated. "BayesPrism" analysis linked higher macrophage abundance to improved survival. A pro-inflammatory FCGR3A + macrophage (Macro_1_FCGR3A) subset exhibited high M1 scores, enhanced antigen presentation, and activation of the PI3K-AKT-mTOR and TNF-NFκB pathways. Pseudotime analysis showed terminal differentiation with elevated HLA-DRB1 and CXCL10. A prognostic model using "StepCox + Ridge" selected 13 key genes, including CX3CR1, achieving AUCs (Area Under the Curve) of 0.77 (training) and 0.69 (validation). CX3CR1 showed strong diagnostic potential (AUC = 0.982). These findings reveal the pivotal role of FCGR3A + macrophages in sepsis prognosis.
{"title":"Integration of single-cell and RNA-seq analysis reveals sepsis heterogeneity and prognostic significance of FCGR3A+ Macrophage subtypes.","authors":"Yijia He, Shuanghu Dong, Lifen Ouyang, Jun Kang, Liying Chen, Qingyun He","doi":"10.1016/j.bbrep.2025.102352","DOIUrl":"10.1016/j.bbrep.2025.102352","url":null,"abstract":"<p><p>Sepsis, a pressing global health challenge, is characterized by immune dysregulation and high mortality. This study, in response to this urgent need, integrated Single-cell RNA sequencing data from 53 samples and bulk transcriptomic data from 479 sepsis patients to analyze immune microenvironment heterogeneity and develop a prognostic signature. Using the scVI algorithm, eight immune cell types were identified. B, T, and NK cells were reduced in sepsis, while neutrophils were elevated. \"BayesPrism\" analysis linked higher macrophage abundance to improved survival. A pro-inflammatory FCGR3A + macrophage (Macro_1_FCGR3A) subset exhibited high M1 scores, enhanced antigen presentation, and activation of the PI3K-AKT-mTOR and TNF-NFκB pathways. Pseudotime analysis showed terminal differentiation with elevated HLA-DRB1 and CXCL10. A prognostic model using \"StepCox + Ridge\" selected 13 key genes, including CX3CR1, achieving AUCs (Area Under the Curve) of 0.77 (training) and 0.69 (validation). CX3CR1 showed strong diagnostic potential (AUC = 0.982). These findings reveal the pivotal role of FCGR3A + macrophages in sepsis prognosis.</p>","PeriodicalId":8771,"journal":{"name":"Biochemistry and Biophysics Reports","volume":"44 ","pages":"102352"},"PeriodicalIF":2.2,"publicationDate":"2025-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12666706/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145660034","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Bikunin is a proteoglycan with a core protein and a low sulfated chondroitin 4-sulfate (Ch4S) chain. This chain has low 4-sulfated glucuronic acid-N-acetylgalactosamine (GlcUAβ1-3GalNAcβ1-4) repeating disaccharide units, and is covalently linked to the core protein through a glucuronic acid-galactose-galactose-xylose-serine the linkage tetrasaccharide (GlcUAβ1-3Galβ1-3Galβ1-4Xylβ1-O-Ser10). The linkage tetrasaccharide is sometimes complexly modified by phosphorylation on Xyl, sulfation on Gal, fucosylation on Xyl, or sialylation on Gal, or combinations of them. Bikunin is used in obstetrics to prevent preterm birth caused by bacterial infection inflammation in Japan. However, the role of the Ch4S chain in the anti-inflammatory action of bikunin has not been fully clarified. To investigate this role, the Ch4S chain was enzymatically removed resulting in a remaining hexasaccharide linkage (GlcUAβ1-3GalNAcβ1-4GlcUAβ1-3Galβ1-3Galβ1-4Xylβ1-O-Ser10) attached to the core peptide. The effects of exogenous bikunin or Ch4S-deficient bikunin on gene expression in lipopolysaccharide (LPS)-treated uterine cervical fibroblasts were comprehensively analyzed by DNA microarray. The expression of IL-8, an inflammation marker in this inflammatory model culture, was upregulated by LPS and the upregulation was inhibited by addition of bikunin together with LPS. Increased expression of genes, related to inflammation or collagen, by LPS was also suppressed by bikunin at the same time point. Ch4S-deficient bikunin did not show significant suppression compared to original bikunin, suggesting that the Ch4S structure of bikunin is important for the suppression of the inflammatory response induced by LPS.
Bikunin是一种具有核心蛋白和低硫酸软骨素4-硫酸(Ch4S)链的蛋白多糖。该链具有低4-硫酸葡萄糖醛酸- n -乙酰半乳糖胺(glcua β1- 3galna β1-4)重复双糖单元,并通过葡萄糖醛酸-半乳糖-半乳糖-木糖-丝氨酸键与核心蛋白共价连接(GlcUAβ1-3Galβ1-3Galβ1-4Xylβ1-O-Ser10)。连接的四糖有时通过在羟基上的磷酸化、在Gal上的硫酸化、在Xyl上的聚焦化或在Gal上的唾液化或它们的组合进行复杂修饰。在日本,Bikunin被用于产科,以防止由细菌感染引起的早产。然而,Ch4S链在比库宁抗炎作用中的作用尚未完全阐明。为了研究这一作用,Ch4S链被酶切去除,导致剩余的六糖链(glcua β1- 3galna β1- 4glcua β1- 3gal β1- 3gal β1- 4xyl β1- o - ser10)附着在核心肽上。采用DNA芯片技术全面分析外源性比库宁和ch4s缺失比库宁对脂多糖(LPS)处理的子宫颈成纤维细胞基因表达的影响。LPS可上调炎症标志物IL-8的表达,比库宁与LPS联合可抑制IL-8的表达。在同一时间点,比库宁也抑制了LPS增加的与炎症或胶原蛋白相关的基因表达。与原比库宁相比,缺乏Ch4S的比库宁没有表现出明显的抑制作用,这表明比库宁的Ch4S结构对抑制LPS诱导的炎症反应很重要。
{"title":"Effect of a chondroitin sulfate chain linked to the bikunin core peptide on gene expression in lipopolysaccharide-treated uterine cervical fibroblasts.","authors":"Saki Kurotaki, Ikuko Kakizaki, Kanji Tanaka, Ryuto Tsushima, Shinichiro Suto, Seigo Tanaka, Tomoe Kodama, Asami Ito, Ryoki Takahashi, Yoshihito Yokoyama","doi":"10.1016/j.bbrep.2025.102363","DOIUrl":"10.1016/j.bbrep.2025.102363","url":null,"abstract":"<p><p>Bikunin is a proteoglycan with a core protein and a low sulfated chondroitin 4-sulfate (Ch4S) chain. This chain has low 4-sulfated glucuronic acid-<i>N</i>-acetylgalactosamine (GlcUAβ1-3GalNAcβ1-4) repeating disaccharide units, and is covalently linked to the core protein through a glucuronic acid-galactose-galactose-xylose-serine the linkage tetrasaccharide (GlcUAβ1-3Galβ1-3Galβ1-4Xylβ1-<i>O</i>-Ser<sup>10</sup>). The linkage tetrasaccharide is sometimes complexly modified by phosphorylation on Xyl, sulfation on Gal, fucosylation on Xyl, or sialylation on Gal, or combinations of them. Bikunin is used in obstetrics to prevent preterm birth caused by bacterial infection inflammation in Japan. However, the role of the Ch4S chain in the anti-inflammatory action of bikunin has not been fully clarified. To investigate this role, the Ch4S chain was enzymatically removed resulting in a remaining hexasaccharide linkage (GlcUAβ1-3GalNAcβ1-4GlcUAβ1-3Galβ1-3Galβ1-4Xylβ1-<i>O</i>-Ser<sup>10</sup>) attached to the core peptide. The effects of exogenous bikunin or Ch4S-deficient bikunin on gene expression in lipopolysaccharide (LPS)-treated uterine cervical fibroblasts were comprehensively analyzed by DNA microarray. The expression of IL-8, an inflammation marker in this inflammatory model culture, was upregulated by LPS and the upregulation was inhibited by addition of bikunin together with LPS. Increased expression of genes, related to inflammation or collagen, by LPS was also suppressed by bikunin at the same time point. Ch4S-deficient bikunin did not show significant suppression compared to original bikunin, suggesting that the Ch4S structure of bikunin is important for the suppression of the inflammatory response induced by LPS.</p>","PeriodicalId":8771,"journal":{"name":"Biochemistry and Biophysics Reports","volume":"44 ","pages":"102363"},"PeriodicalIF":2.2,"publicationDate":"2025-11-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12666056/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145660067","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-17eCollection Date: 2025-12-01DOI: 10.1016/j.bbrep.2025.102367
Yuqin Wei, Xingwen Wei, Wei Zhao
Objective: This study aimed to investigate the prognostic value and biological implications of cuproptosis/ferroptosis-related genes in breast cancer, and to develop a robust molecular signature for risk stratification and treatment guidance.
Methods: Integrative bioinformatic analysis of TCGA and GEO datasets identified cuproptosis/ferroptosis-related genes. LASSO and Cox regression were used to refine a multi-gene signature. Unsupervised clustering stratified molecular subtypes. Genomic alterations, immune microenvironment composition, and drug sensitivity were assessed using bioinformatic tools. Experimental validation included immunohistochemistry and Western blot using clinical samples and cell lines (MCF-10A, MDA-MB-231, BT-549, SKBR-3, MCF-7).
Results: A four-gene signature (MTDH, PROM2, IFNG, SLC1A4) with independent prognostic value was established. Subtype-specific expression patterns emerged: IFNG associated with basal-like tumors and cytotoxic immunity, while SLC1A4 (HER2-enriched/luminal) and MTDH/PROM2 correlated with immunosuppressive microenvironments. Disease progression showed stage-dependent attenuation of IFNG/MTDH/PROM2 and progressive SLC1A4 upregulation. Genomic profiling revealed recurrent mutations and frequent copy-number amplifications, notably of MTDH (60.37 %) and loci on chromosomes 2/3. Functional assays demonstrated IFNG/MTDH/SLC1A4-mediated resistance to anthracyclines and microtubule inhibitors, contrasting with PROM2-associated chemosensitivity. Unsupervised clustering identified two molecular subtypes with divergent survival, characterized by distinct pathway activation. Prognostic nomograms achieved robust predictive accuracy (C-index: 0.755). Validation confirmed upregulation of all four genes in breast cancer.
Conclusion: The cuproptosis/ferroptosis-related genes-based signature serves as a reliable prognostic tool, reflecting immune landscape remodeling and genomic instability in BRCA. These findings provide insights into subtype-specific therapeutic vulnerabilities and suggest potential strategies for targeting cuproptosis/ferroptosis pathways in precision oncology.
{"title":"Integrated bioinformatics analysis identifies cuproptosis and ferroptosis gene signatures via subgroup clustering: Prognostic and therapeutic implications in breast cancer.","authors":"Yuqin Wei, Xingwen Wei, Wei Zhao","doi":"10.1016/j.bbrep.2025.102367","DOIUrl":"10.1016/j.bbrep.2025.102367","url":null,"abstract":"<p><strong>Objective: </strong>This study aimed to investigate the prognostic value and biological implications of cuproptosis/ferroptosis-related genes in breast cancer, and to develop a robust molecular signature for risk stratification and treatment guidance.</p><p><strong>Methods: </strong>Integrative bioinformatic analysis of TCGA and GEO datasets identified cuproptosis/ferroptosis-related genes. LASSO and Cox regression were used to refine a multi-gene signature. Unsupervised clustering stratified molecular subtypes. Genomic alterations, immune microenvironment composition, and drug sensitivity were assessed using bioinformatic tools. Experimental validation included immunohistochemistry and Western blot using clinical samples and cell lines (MCF-10A, MDA-MB-231, BT-549, SKBR-3, MCF-7).</p><p><strong>Results: </strong>A four-gene signature (MTDH, PROM2, IFNG, SLC1A4) with independent prognostic value was established. Subtype-specific expression patterns emerged: IFNG associated with basal-like tumors and cytotoxic immunity, while SLC1A4 (HER2-enriched/luminal) and MTDH/PROM2 correlated with immunosuppressive microenvironments. Disease progression showed stage-dependent attenuation of IFNG/MTDH/PROM2 and progressive SLC1A4 upregulation. Genomic profiling revealed recurrent mutations and frequent copy-number amplifications, notably of MTDH (60.37 %) and loci on chromosomes 2/3. Functional assays demonstrated IFNG/MTDH/SLC1A4-mediated resistance to anthracyclines and microtubule inhibitors, contrasting with PROM2-associated chemosensitivity. Unsupervised clustering identified two molecular subtypes with divergent survival, characterized by distinct pathway activation. Prognostic nomograms achieved robust predictive accuracy (C-index: 0.755). Validation confirmed upregulation of all four genes in breast cancer.</p><p><strong>Conclusion: </strong>The cuproptosis/ferroptosis-related genes-based signature serves as a reliable prognostic tool, reflecting immune landscape remodeling and genomic instability in BRCA. These findings provide insights into subtype-specific therapeutic vulnerabilities and suggest potential strategies for targeting cuproptosis/ferroptosis pathways in precision oncology.</p>","PeriodicalId":8771,"journal":{"name":"Biochemistry and Biophysics Reports","volume":"44 ","pages":"102367"},"PeriodicalIF":2.2,"publicationDate":"2025-11-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12666366/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145660018","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-17eCollection Date: 2025-12-01DOI: 10.1016/j.bbrep.2025.102366
Xianxiang Chen, Qingle Zeng, Min Xia, Yufen Chen
Aims: Individuals with diabetic nephropathy (DN), a major diabetic complication, have been disproportionately affected by the coronavirus disease 2019 (COVID-19) pandemic. This study aimed to investigate the molecular interplay between COVID-19 and DN using bioinformatics and systems biology approaches to identify shared mechanisms and therapeutic targets for their improved synergistic clinical management.
Methods: Transcriptomic datasets (COVID-19, GSE171110; DN, GSE30528) were analyzed to identify differentially expressed genes (DEGs). Additionally, functional enrichment, protein-protein interaction (PPI) networks, transcription factor (TF)-microRNA (miRNA) regulatory networks, and drug-gene associations were explored. The diagnostic potential of hub genes was validated using receiver operating characteristic curves.
Results: In total, 3975 DEGs (2796 upregulated; 1179 downregulated) were identified in patients with COVID-19 versus controls, and 348 DEGs (93 upregulated; 255 downregulated) were found in patients with DN. Among them, 83 DEGs overlapped, presenting shared molecular pathways, including hematopoietic cell lineage, focal adhesion, and complement/coagulation cascades. PPI analysis revealed five major hub genes (IL7R, CD2, GZMA, CD3D, and FCER1A) associated with immune regulation and tissue injury, and regulatory network analysis identified 46 TFs and 88 miRNAs interacting with them. Based on transcriptomic signatures, drug repurposing candidates, such as alpha-d-mannose, aspirin, and methotrexate, were identified. Additionally, hub genes showed a high diagnostic potential (area under the curve >0.80 for COVID-19 and DN). Finally, we use external datasets to validate hub genes.
Conclusions: The findings of this study reveal shared molecular pathways and hub genes between COVID-19 and DN, providing insights into immune dysregulation and tissue injury mechanisms. Strategies associated with identified biomarkers and therapeutic candidates, including interleukin-7 receptor-targeting strategies, offer the potential for improving clinical outcomes in patients with comorbid COVID-19 and DN. Lastly, these findings underscore the value of integrative bioinformatics in guiding precision medicine approaches for complex disease interactions.
{"title":"Decoding the interplay between COVID-19 and diabetic nephropathy through bioinformatics and systems biology techniques.","authors":"Xianxiang Chen, Qingle Zeng, Min Xia, Yufen Chen","doi":"10.1016/j.bbrep.2025.102366","DOIUrl":"10.1016/j.bbrep.2025.102366","url":null,"abstract":"<p><strong>Aims: </strong>Individuals with diabetic nephropathy (DN), a major diabetic complication, have been disproportionately affected by the coronavirus disease 2019 (COVID-19) pandemic. This study aimed to investigate the molecular interplay between COVID-19 and DN using bioinformatics and systems biology approaches to identify shared mechanisms and therapeutic targets for their improved synergistic clinical management.</p><p><strong>Methods: </strong>Transcriptomic datasets (COVID-19, GSE171110; DN, GSE30528) were analyzed to identify differentially expressed genes (DEGs). Additionally, functional enrichment, protein-protein interaction (PPI) networks, transcription factor (TF)-microRNA (miRNA) regulatory networks, and drug-gene associations were explored. The diagnostic potential of hub genes was validated using receiver operating characteristic curves.</p><p><strong>Results: </strong>In total, 3975 DEGs (2796 upregulated; 1179 downregulated) were identified in patients with COVID-19 versus controls, and 348 DEGs (93 upregulated; 255 downregulated) were found in patients with DN. Among them, 83 DEGs overlapped, presenting shared molecular pathways, including hematopoietic cell lineage, focal adhesion, and complement/coagulation cascades. PPI analysis revealed five major hub genes (<i>IL7R</i>, <i>CD2</i>, <i>GZMA</i>, <i>CD3D</i>, and <i>FCER1A</i>) associated with immune regulation and tissue injury, and regulatory network analysis identified 46 TFs and 88 miRNAs interacting with them. Based on transcriptomic signatures, drug repurposing candidates, such as alpha-d-mannose, aspirin, and methotrexate, were identified. Additionally, hub genes showed a high diagnostic potential (area under the curve >0.80 for COVID-19 and DN). Finally, we use external datasets to validate hub genes.</p><p><strong>Conclusions: </strong>The findings of this study reveal shared molecular pathways and hub genes between COVID-19 and DN, providing insights into immune dysregulation and tissue injury mechanisms. Strategies associated with identified biomarkers and therapeutic candidates, including interleukin-7 receptor-targeting strategies, offer the potential for improving clinical outcomes in patients with comorbid COVID-19 and DN. Lastly, these findings underscore the value of integrative bioinformatics in guiding precision medicine approaches for complex disease interactions.</p>","PeriodicalId":8771,"journal":{"name":"Biochemistry and Biophysics Reports","volume":"44 ","pages":"102366"},"PeriodicalIF":2.2,"publicationDate":"2025-11-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12666057/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145660024","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-17eCollection Date: 2025-12-01DOI: 10.1016/j.bbrep.2025.102357
Farooqahmed S Kittur, Finn Hynes, Naasir Albright, Qaisar Mahmood, Jiahua Xie
Erythropoietin (Epo) is a glycoprotein hormone primarily known for regulating red blood cell production. Beyond erythropoiesis, Epo has been reported to exert tissue-protective effects, including neuroprotective and neuroregenerative properties. While its erythropoietic activity is mediated by the Epo receptor (EpoR), its tissue-protective function has been suggested to involve alternative receptor(s), the existence of which remains unclear. To investigate the effects of Epo on neuronal cell growth and explore potential tissue-protective receptors in the future, we stably expressed the human EPO gene in the mouse neuroblastoma cell line Neuro-2A (N2A) and hippocampal neuronal cell line HT22 and investigated the effects of Epo overexpression on neuronal cell proliferation. ELISA and Western blot analyses confirmed successful Epo overexpression in both cell lines. Notably, N2A cells overexpressing Epo (N2A+Epo) exhibited a rapid increase in cell number compared to vector-only controls, suggesting Epo-stimulated cell proliferation. Consistent with this observation, N2A+Epo cells showed increased BrdU incorporation and elevated levels of cell proliferation markers, including proliferating cell nuclear antigen (PCNA) and nucleophosmin (NPM1), as revealed by Western blot analysis, compared to the vector-only controls. These findings indicate increased DNA synthesis and cell proliferation in N2A+Epo cells. Additionally, CRISPR-Cas9-mediated knockdown of EPO resulted in the abrogation of cell proliferation, whereas EPOR knockdown showed no effect, suggesting that Epo promotes N2A cell proliferation through an EpoR-independent mechanism. Collectively, these findings highlight the potential of the N2A+Epo cell line as a model for identifying alternative tissue-protective Epo receptors.
{"title":"Erythropoietin promotes cell proliferation in Neuro-2A mouse neuroblastoma cells.","authors":"Farooqahmed S Kittur, Finn Hynes, Naasir Albright, Qaisar Mahmood, Jiahua Xie","doi":"10.1016/j.bbrep.2025.102357","DOIUrl":"10.1016/j.bbrep.2025.102357","url":null,"abstract":"<p><p>Erythropoietin (Epo) is a glycoprotein hormone primarily known for regulating red blood cell production. Beyond erythropoiesis, Epo has been reported to exert tissue-protective effects, including neuroprotective and neuroregenerative properties. While its erythropoietic activity is mediated by the Epo receptor (EpoR), its tissue-protective function has been suggested to involve alternative receptor(s), the existence of which remains unclear. To investigate the effects of Epo on neuronal cell growth and explore potential tissue-protective receptors in the future, we stably expressed the human <i>EPO</i> gene in the mouse neuroblastoma cell line Neuro-2A (N2A) and hippocampal neuronal cell line HT22 and investigated the effects of Epo overexpression on neuronal cell proliferation. ELISA and Western blot analyses confirmed successful Epo overexpression in both cell lines. Notably, N2A cells overexpressing Epo (N2A<sup>+Epo</sup>) exhibited a rapid increase in cell number compared to vector-only controls, suggesting Epo-stimulated cell proliferation. Consistent with this observation, N2A<sup>+Epo</sup> cells showed increased BrdU incorporation and elevated levels of cell proliferation markers, including proliferating cell nuclear antigen (PCNA) and nucleophosmin (NPM1), as revealed by Western blot analysis, compared to the vector-only controls. These findings indicate increased DNA synthesis and cell proliferation in N2A<sup>+Epo</sup> cells. Additionally, CRISPR-Cas9-mediated knockdown of <i>EPO</i> resulted in the abrogation of cell proliferation, whereas <i>EPOR</i> knockdown showed no effect, suggesting that Epo promotes N2A cell proliferation through an EpoR-independent mechanism. Collectively, these findings highlight the potential of the N2A<sup>+Epo</sup> cell line as a model for identifying alternative tissue-protective Epo receptors.</p>","PeriodicalId":8771,"journal":{"name":"Biochemistry and Biophysics Reports","volume":"44 ","pages":"102357"},"PeriodicalIF":2.2,"publicationDate":"2025-11-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12664986/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145653433","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-17eCollection Date: 2025-12-01DOI: 10.1016/j.bbrep.2025.102361
Zheng Zhao, Lulu Lu, Yang Yi, Na Gao, Jiangang Hu, Gangwen Han, Xiaolei Ma
<p><strong>Background: </strong>Rosacea is a chronic inflammatory skin disease characterized by diverse symptoms and variable clinical progression, which can significantly impair patients' quality of life and mental health. The exact etiology of rosacea remains elusive. It has been hypothesized that specific microorganisms may trigger symptom onset and play crucial roles in the pathogenesis of the disease.</p><p><strong>Objective: </strong>We performed a case-control study to investigate the gut microbiome of rosacea patients compared to controls matched by age, sex in China.</p><p><strong>Methods: </strong>The study cohort comprised eight patients diagnosed with rosacea and eight age- and sex-matched healthy controls residing in Beijing. Metagenomic sequencing was performed using on a llumina Novaseq 6000 platform. Hospital Anxiety and Depression Scale was used to evaluate the severity of anxiety and depression of rosacea patients. Skindex-16 score was used to assess dermatology-specific health-related quality of life (HrQoL) in patients with rosacea. The clinical evaluation of acne was done using the ECLA score.</p><p><strong>Results: </strong>The rosacea patients showed higher HADS and Skindex-16 score (15.375 ± 1.302 and 46 ± 9.75 respectively) vs healthy controls (3.425 ± 1.308 and 0 respectively). A clear distinction was observed between the rosacea group and the control group, characterized by a significant increase in the abundance of Turicibacter_sp._TJ11, Turicibacter_sp._H121,Turicibacter_sp._TA25,Turicibacter_sp._T129<b>,</b>Ruminococcus_sp._AF18-22,Ruminococcus_sp._CAG:379,Ruminococcus_sp._AM2829LB,Ruminococcus_callidus, Ruminococcus_sp._AM36-18,Ruminococcus_sp._AF43-11,Ruminococcus_sp._AM28-41,Streptococcus sp. 23.2,Streptococcus infantarius, Streptococcus vestibularis, Streptococcus salivarius, Streptococcus gordonii, Clostridium_sp._CAG:798, Clostridium_tertium, Alistipes_sp._Z76 and Lachnospiraceae_bacterium_XBB2008in the rosacea group. In contrast, reduced levels were detected in the rosacea group for Clostridium_sp._AF12-41, Clostridium_sp._CAG:299, Clostridium_sp._OM05-5BH,Clostridium_sp._AF24-2LB, Clostridium_sp._AM18-55, Clostridium_sp._CAG:43, Clostridium_sp._OM047,Clostridium_sp._TF1113AC,Clostridium_sp._OF134,Clostridium_disporicum, Butyrivibrio_sp._CB08,Butyrivibrio_sp._INlla14<b>,</b> Roseburia_sp._CAG:50 (p < 0.05). Pearson correlation analysis revealed that Gemmiger_sp._An120 was positively correlated with Skindex-16 and negatively correlated with ECLA score (P < 0.05). Clostridium_sp._CAG:299 was negatively correlated with HADS scores and positive correlation with ECLA score (P < 0.05). KEGG pathway analysis found KO05034, KO04024 and KO00920 pathways exhibited increased activity in the Rosacea group (P < 0.05).</p><p><strong>Conclusions: </strong>The gut microbiota in individuals with rosacea displayed changed from that of healthy control. These microbial alterations may contribute to the pathogenesis of rosacea thr
{"title":"Gut microbiota signature in a cohort of Chinese patients with rosacea.","authors":"Zheng Zhao, Lulu Lu, Yang Yi, Na Gao, Jiangang Hu, Gangwen Han, Xiaolei Ma","doi":"10.1016/j.bbrep.2025.102361","DOIUrl":"10.1016/j.bbrep.2025.102361","url":null,"abstract":"<p><strong>Background: </strong>Rosacea is a chronic inflammatory skin disease characterized by diverse symptoms and variable clinical progression, which can significantly impair patients' quality of life and mental health. The exact etiology of rosacea remains elusive. It has been hypothesized that specific microorganisms may trigger symptom onset and play crucial roles in the pathogenesis of the disease.</p><p><strong>Objective: </strong>We performed a case-control study to investigate the gut microbiome of rosacea patients compared to controls matched by age, sex in China.</p><p><strong>Methods: </strong>The study cohort comprised eight patients diagnosed with rosacea and eight age- and sex-matched healthy controls residing in Beijing. Metagenomic sequencing was performed using on a llumina Novaseq 6000 platform. Hospital Anxiety and Depression Scale was used to evaluate the severity of anxiety and depression of rosacea patients. Skindex-16 score was used to assess dermatology-specific health-related quality of life (HrQoL) in patients with rosacea. The clinical evaluation of acne was done using the ECLA score.</p><p><strong>Results: </strong>The rosacea patients showed higher HADS and Skindex-16 score (15.375 ± 1.302 and 46 ± 9.75 respectively) vs healthy controls (3.425 ± 1.308 and 0 respectively). A clear distinction was observed between the rosacea group and the control group, characterized by a significant increase in the abundance of Turicibacter_sp._TJ11, Turicibacter_sp._H121,Turicibacter_sp._TA25,Turicibacter_sp._T129<b>,</b>Ruminococcus_sp._AF18-22,Ruminococcus_sp._CAG:379,Ruminococcus_sp._AM2829LB,Ruminococcus_callidus, Ruminococcus_sp._AM36-18,Ruminococcus_sp._AF43-11,Ruminococcus_sp._AM28-41,Streptococcus sp. 23.2,Streptococcus infantarius, Streptococcus vestibularis, Streptococcus salivarius, Streptococcus gordonii, Clostridium_sp._CAG:798, Clostridium_tertium, Alistipes_sp._Z76 and Lachnospiraceae_bacterium_XBB2008in the rosacea group. In contrast, reduced levels were detected in the rosacea group for Clostridium_sp._AF12-41, Clostridium_sp._CAG:299, Clostridium_sp._OM05-5BH,Clostridium_sp._AF24-2LB, Clostridium_sp._AM18-55, Clostridium_sp._CAG:43, Clostridium_sp._OM047,Clostridium_sp._TF1113AC,Clostridium_sp._OF134,Clostridium_disporicum, Butyrivibrio_sp._CB08,Butyrivibrio_sp._INlla14<b>,</b> Roseburia_sp._CAG:50 (p < 0.05). Pearson correlation analysis revealed that Gemmiger_sp._An120 was positively correlated with Skindex-16 and negatively correlated with ECLA score (P < 0.05). Clostridium_sp._CAG:299 was negatively correlated with HADS scores and positive correlation with ECLA score (P < 0.05). KEGG pathway analysis found KO05034, KO04024 and KO00920 pathways exhibited increased activity in the Rosacea group (P < 0.05).</p><p><strong>Conclusions: </strong>The gut microbiota in individuals with rosacea displayed changed from that of healthy control. These microbial alterations may contribute to the pathogenesis of rosacea thr","PeriodicalId":8771,"journal":{"name":"Biochemistry and Biophysics Reports","volume":"44 ","pages":"102361"},"PeriodicalIF":2.2,"publicationDate":"2025-11-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12666052/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145660049","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-16DOI: 10.1016/j.bbrep.2025.102371
Ye Qiu , Xingying Zhu , Xiaoqian Yang , Jiaming Wan
Skin wound healing is a complex and highly coordinated biological process involving multiple cell types and signaling pathways, among which fibroblasts play a pivotal role. Exosomes, as nanoscale extracellular vesicles that mediate intercellular communication, have emerged as critical regulators in tissue repair. This review summarizes the biological characteristics of fibroblast-derived exosomes and their regulatory roles and molecular mechanisms in skin wound healing. Fibroblast exosomes accelerate wound closure by promoting cell proliferation and migration, modulating inflammatory responses, enhancing angiogenesis, and remodeling the extracellular matrix (ECM). Their cargo—comprising bioactive molecules such as microRNAs (miRNAs), proteins, and growth factors—participates in multiple signaling pathways including TGF-β/Smad, PI3K/Akt, and NF-κB, thereby orchestrating cellular behaviors essential for effective tissue regeneration. Specific miRNAs, such as miR-21, miR-29, and miR-146a, have been identified as key mediators regulating fibroblast differentiation, ECM synthesis, and inflammatory resolution. Furthermore, fibroblast exosomes exhibit considerable potential in clinical applications, including chronic wound treatment, tissue engineering, regenerative medicine, and drug delivery systems, owing to their low immunogenicity and intrinsic bioactivity. Despite these advances, challenges remain in the standardization, large-scale production, and safety evaluation of exosome-based therapies. Future studies focusing on elucidating exosome heterogeneity, optimizing biomanufacturing processes, and ensuring clinical safety will be crucial to translating fibroblast-derived exosomes into effective therapeutic agents. Collectively, fibroblast exosomes represent a promising cell-free strategy for promoting wound healing and advancing regenerative medicine.
{"title":"Mechanisms of skin wound healing regulated by fibroblast-derived exosomes","authors":"Ye Qiu , Xingying Zhu , Xiaoqian Yang , Jiaming Wan","doi":"10.1016/j.bbrep.2025.102371","DOIUrl":"10.1016/j.bbrep.2025.102371","url":null,"abstract":"<div><div>Skin wound healing is a complex and highly coordinated biological process involving multiple cell types and signaling pathways, among which fibroblasts play a pivotal role. Exosomes, as nanoscale extracellular vesicles that mediate intercellular communication, have emerged as critical regulators in tissue repair. This review summarizes the biological characteristics of fibroblast-derived exosomes and their regulatory roles and molecular mechanisms in skin wound healing. Fibroblast exosomes accelerate wound closure by promoting cell proliferation and migration, modulating inflammatory responses, enhancing angiogenesis, and remodeling the extracellular matrix (ECM). Their cargo—comprising bioactive molecules such as microRNAs (miRNAs), proteins, and growth factors—participates in multiple signaling pathways including TGF-β/Smad, PI3K/Akt, and NF-κB, thereby orchestrating cellular behaviors essential for effective tissue regeneration. Specific miRNAs, such as miR-21, miR-29, and miR-146a, have been identified as key mediators regulating fibroblast differentiation, ECM synthesis, and inflammatory resolution. Furthermore, fibroblast exosomes exhibit considerable potential in clinical applications, including chronic wound treatment, tissue engineering, regenerative medicine, and drug delivery systems, owing to their low immunogenicity and intrinsic bioactivity. Despite these advances, challenges remain in the standardization, large-scale production, and safety evaluation of exosome-based therapies. Future studies focusing on elucidating exosome heterogeneity, optimizing biomanufacturing processes, and ensuring clinical safety will be crucial to translating fibroblast-derived exosomes into effective therapeutic agents. Collectively, fibroblast exosomes represent a promising cell-free strategy for promoting wound healing and advancing regenerative medicine.</div></div>","PeriodicalId":8771,"journal":{"name":"Biochemistry and Biophysics Reports","volume":"44 ","pages":"Article 102371"},"PeriodicalIF":2.2,"publicationDate":"2025-11-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145576361","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-16eCollection Date: 2025-12-01DOI: 10.1016/j.bbrep.2025.102365
Natalie G Horgan, Anabela Djurovic-Topalovic, Taiwo A Ademoye, Hannah I Reyes-Charles, Natsumi Kobayashi, Germán Plascencia-Villa, George Perry, Tomoaki Murakami, Jessica S Fortin
Serum amyloid A1 (SAA1) is a 122-amino acid protein that, after cleavage, matures into a 104-amino acid form. Its N-terminus is responsible for binding high-density lipoprotein (HDL), while the C-terminus maintains its structural integrity. As an acute-phase protein, SAA1 is produced by the liver in response to acute inflammation. SAA1 is also a precursor to amyloid A (AA), and its accumulation can lead to AA amyloidosis-a condition secondary to chronic inflammation that causes tissue damage and organ dysfunction. Our study explores methods to disaggregate SAA1 fibrils isolated from the cat spleen, chicken liver, and cow liver. Specifically, we investigate the use of epigallocatechin-3-gallate (EGCG), a polyphenolic flavonoid extracted from green tea known for its anti-inflammatory and antioxidant properties, to disaggregate these fibrils. Dynamic light scattering (DLS) and transmission electron microscopy (TEM) were used to analyze these fibrils after treatment with 1 % DMSO and 400 μM of EGCG in 10 mM PBS (pH 7.4). The results demonstrated that EGCG effectively reduced fibril size, as confirmed by DLS characterization, with the disappearance or diminished prominence of the 103-4 nm peak. Additional TEM results confirmed that EGCG disaggregated amyloid-beta fibrils isolated from Alzheimer's disease brains. These findings suggest that compounds like EGCG could be valuable in treating inflammatory and neurodegenerative conditions by disaggregating amyloid fibrils.
{"title":"Epigallocatechin-3-Gallate: A potential amyloid Fibril Disaggregator of Serum amyloid A1.","authors":"Natalie G Horgan, Anabela Djurovic-Topalovic, Taiwo A Ademoye, Hannah I Reyes-Charles, Natsumi Kobayashi, Germán Plascencia-Villa, George Perry, Tomoaki Murakami, Jessica S Fortin","doi":"10.1016/j.bbrep.2025.102365","DOIUrl":"10.1016/j.bbrep.2025.102365","url":null,"abstract":"<p><p>Serum amyloid A1 (SAA1) is a 122-amino acid protein that, after cleavage, matures into a 104-amino acid form. Its N-terminus is responsible for binding high-density lipoprotein (HDL), while the C-terminus maintains its structural integrity. As an acute-phase protein, SAA1 is produced by the liver in response to acute inflammation. SAA1 is also a precursor to amyloid A (AA), and its accumulation can lead to AA amyloidosis-a condition secondary to chronic inflammation that causes tissue damage and organ dysfunction. Our study explores methods to disaggregate SAA1 fibrils isolated from the cat spleen, chicken liver, and cow liver. Specifically, we investigate the use of epigallocatechin-3-gallate (EGCG), a polyphenolic flavonoid extracted from green tea known for its anti-inflammatory and antioxidant properties, to disaggregate these fibrils. Dynamic light scattering (DLS) and transmission electron microscopy (TEM) were used to analyze these fibrils after treatment with 1 % DMSO and 400 μM of EGCG in 10 mM PBS (pH 7.4). The results demonstrated that EGCG effectively reduced fibril size, as confirmed by DLS characterization, with the disappearance or diminished prominence of the 10<sup>3-4</sup> nm peak. Additional TEM results confirmed that EGCG disaggregated amyloid-beta fibrils isolated from Alzheimer's disease brains. These findings suggest that compounds like EGCG could be valuable in treating inflammatory and neurodegenerative conditions by disaggregating amyloid fibrils.</p>","PeriodicalId":8771,"journal":{"name":"Biochemistry and Biophysics Reports","volume":"44 ","pages":"102365"},"PeriodicalIF":2.2,"publicationDate":"2025-11-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12664600/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145647171","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-15eCollection Date: 2025-12-01DOI: 10.1016/j.bbrep.2025.102325
Jingbo Li, Junjie Wang, Yinian Lu, Yan Wang, Yang Cao, Yuanyuan Li, Yinglin Cui
Allergic rhinitis (AR) is a chronic inflammatory disease with complex causes. Traditional Chinese medicine (TCM) shows promise for treating AR. This study aimed to explore the mechanisms behind TCM's effectiveness. In this study, we collected peripheral blood mononuclear cells (PBMCs) from AR patients before and after TCM treatment. Single-cell RNA sequencing (scRNA-seq) was performed to identify cell types and differentially expressed genes (DEGs) and analyzed immune cell pathways to understand how TCM treats AR. Cell-cell communication and regulatory networks were also investigated. Accordingly, 11 major cell types were annotated. The strength of the outgoing interaction between CD14+ monocytes and Central Memory CD4+ T cells and Transitional B cells decreased and in-creased, respectively. Transcription factors (TFs), VDR, and RUNX3 regulated the balance of Th1/Th2 cells to improve AR symptoms. This study confirms the therapeutic effect of Peiyuan Tongqiao Decoction on AR and provides insights into its mechanism of action.
{"title":"Single-cell RNA sequencing reveals immune cell alterations in patients with allergic rhinitis treated with Peiyuan Tong-qiao decoction.","authors":"Jingbo Li, Junjie Wang, Yinian Lu, Yan Wang, Yang Cao, Yuanyuan Li, Yinglin Cui","doi":"10.1016/j.bbrep.2025.102325","DOIUrl":"10.1016/j.bbrep.2025.102325","url":null,"abstract":"<p><p>Allergic rhinitis (AR) is a chronic inflammatory disease with complex causes. Traditional Chinese medicine (TCM) shows promise for treating AR. This study aimed to explore the mechanisms behind TCM's effectiveness. In this study, we collected peripheral blood mononuclear cells (PBMCs) from AR patients before and after TCM treatment. Single-cell RNA sequencing (scRNA-seq) was performed to identify cell types and differentially expressed genes (DEGs) and analyzed immune cell pathways to understand how TCM treats AR. Cell-cell communication and regulatory networks were also investigated. Accordingly, 11 major cell types were annotated. The strength of the outgoing interaction between CD14<sup>+</sup> monocytes and Central Memory CD4<sup>+</sup> T cells and Transitional B cells decreased and in-creased, respectively. Transcription factors (TFs), VDR, and RUNX3 regulated the balance of Th1/Th2 cells to improve AR symptoms. This study confirms the therapeutic effect of Peiyuan Tongqiao Decoction on AR and provides insights into its mechanism of action.</p>","PeriodicalId":8771,"journal":{"name":"Biochemistry and Biophysics Reports","volume":"44 ","pages":"102325"},"PeriodicalIF":2.2,"publicationDate":"2025-11-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12663851/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145647192","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}