Pub Date : 2026-01-01DOI: 10.1016/j.csbj.2026.01.002
Sung-Yeon Lee, Seongwon Ma, Sangjun Davie Jeon, Hyoju Kim, Beomjoon Jo, Seung-Hoon Han, Eunsoo Jang, Jimin Lee, Yong-Kyu Lee, Dasom Lee
CRISPR/Cas9 has transformed gene editing, enabling precise genetic modifications across species. However, existing sgRNA design prediction models based on in vitro data are difficult to generalize to in vivo contexts. In particular, approaches based on single-sgRNA design require additional filtering of in-frame mutations, which is inefficient in terms of both time and cost. In this study, we developed the first mammalian in vivo-trained prediction model to evaluate the efficiency of a dual-sgRNA-based exon deletion strategy. Using 230 editing outcomes of postnatal viable individuals, eight prediction models were constructed and evaluated based on generalized linear models and Random Forests. The final selected model, a Combined GLM, integrated the DeepSpCas9 score with k-mer sequence features, achieving an AUC of 0.759 (95 % Confidence Interval: 0.697–0.821). Motif analysis revealed that CC sequences were associated with high efficiency and TT sequences were associated with low editing efficiency. This study demonstrates that integrating sequence-based features with existing design scores can improve sgRNA efficiency prediction in vivo. The proposed framework can be applied to the development of next-generation sgRNA design tools, with implications for gene therapy, effective animal model generation, and precision genome engineering.
{"title":"Empirical optimization of dual-sgRNA design for in vivo CRISPR/Cas9-mediated exon deletion in mice","authors":"Sung-Yeon Lee, Seongwon Ma, Sangjun Davie Jeon, Hyoju Kim, Beomjoon Jo, Seung-Hoon Han, Eunsoo Jang, Jimin Lee, Yong-Kyu Lee, Dasom Lee","doi":"10.1016/j.csbj.2026.01.002","DOIUrl":"10.1016/j.csbj.2026.01.002","url":null,"abstract":"<div><div>CRISPR/Cas9 has transformed gene editing, enabling precise genetic modifications across species. However, existing sgRNA design prediction models based on in vitro data are difficult to generalize to in vivo contexts. In particular, approaches based on single-sgRNA design require additional filtering of in-frame mutations, which is inefficient in terms of both time and cost. In this study, we developed the first mammalian in vivo-trained prediction model to evaluate the efficiency of a dual-sgRNA-based exon deletion strategy. Using 230 editing outcomes of postnatal viable individuals, eight prediction models were constructed and evaluated based on generalized linear models and Random Forests. The final selected model, a Combined GLM, integrated the DeepSpCas9 score with k-mer sequence features, achieving an AUC of 0.759 (95 % Confidence Interval: 0.697–0.821). Motif analysis revealed that CC sequences were associated with high efficiency and TT sequences were associated with low editing efficiency. This study demonstrates that integrating sequence-based features with existing design scores can improve sgRNA efficiency prediction in vivo. The proposed framework can be applied to the development of next-generation sgRNA design tools, with implications for gene therapy, effective animal model generation, and precision genome engineering.</div></div>","PeriodicalId":10715,"journal":{"name":"Computational and structural biotechnology journal","volume":"31 ","pages":"Pages 355-362"},"PeriodicalIF":4.1,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145921289","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-01DOI: 10.1016/j.csbj.2025.12.023
Silvia Tapia-Gonzalez , Josué García Yagüe , George E. Barreto
Major depressive disorder (MDD) is a multifactorial mental health condition involving genetic, environmental, and neurobiological factors. Conventional antidepressants such as fluoxetine, a selective serotonin reuptake inhibitor, require weeks to exert therapeutic effects, whereas ketamine and esketamine act rapidly via glutamatergic modulation. These drugs may also converge on the inhibition of glycogen synthase kinase 3 beta (GSK3B) as a key mechanism for their antidepressant effects, increasing neuroplasticity, synaptic transmission, and neuronal survival through upregulation of brain-derived neurotrophic factor (BDNF). Part of the antidepressant effects of ketamine also seems to depend on opioid receptor activation. Despite recent progress, variability in antidepressant response in MDD remains unclear. This work explores, via meta-analysis and network fragility analysis, key molecular mechanisms in MDD, how these drugs exert actions, and highlights potential therapeutic targets for MDD. We performed a network pharmacology approach to unravel the key cellular processes involved in MDD, including altered synaptic plasticity, neurogenesis, apoptosis, and neuroinflammation. Second, we explored the therapeutic role of these treatments on these altered cellular processes. By integrating drug-target data with MDD-associated genes, we identified the opioid receptor mu 1 (OPRM1), epidermal growth factor receptor (EGFR) and GSK3B as key druggable targets. Network analysis further suggested that nuclear factor kappa B (NFKB) may regulate all three, positioning it as a central node linking inflammation, synaptic plasticity, and neuronal metabolism in MDD. We hypothesize that targeted modulation of these genes may optimize the therapeutic efficacy, while NFKB emerges as a promising candidate biomarker for guiding treatment strategies in MDD.
重度抑郁症(MDD)是一种涉及遗传、环境和神经生物学因素的多因素精神健康状况。传统的抗抑郁药,如氟西汀,一种选择性血清素再摄取抑制剂,需要数周才能发挥治疗效果,而氯胺酮和艾氯胺酮通过谷氨酸调节迅速起作用。这些药物也可能集中于抑制糖原合成酶激酶3 β (GSK3B),这是其抗抑郁作用的关键机制,通过上调脑源性神经营养因子(BDNF)来增加神经可塑性、突触传递和神经元存活。氯胺酮的部分抗抑郁作用似乎也依赖于阿片受体的激活。尽管最近取得了进展,但抑郁症患者抗抑郁反应的变异性仍不清楚。本研究通过荟萃分析和网络脆弱性分析,探讨了MDD的关键分子机制,这些药物如何发挥作用,并强调了MDD的潜在治疗靶点。我们采用网络药理学方法来揭示与MDD相关的关键细胞过程,包括突触可塑性改变、神经发生、细胞凋亡和神经炎症。其次,我们探索了这些治疗对这些改变的细胞过程的治疗作用。通过整合药物靶点数据和mdd相关基因,我们确定了阿片受体mu 1 (OPRM1)、表皮生长因子受体(EGFR)和GSK3B作为关键的可药物靶点。网络分析进一步表明,核因子κ B (NFKB)可能调节这三者,将其定位为MDD中连接炎症、突触可塑性和神经元代谢的中心节点。我们假设这些基因的靶向调节可能会优化治疗效果,而NFKB则成为指导MDD治疗策略的有希望的候选生物标志物。
{"title":"Network pharmacology of cellular targets in major depressive disorder and differential mechanisms of fluoxetine, ketamine and esketamine","authors":"Silvia Tapia-Gonzalez , Josué García Yagüe , George E. Barreto","doi":"10.1016/j.csbj.2025.12.023","DOIUrl":"10.1016/j.csbj.2025.12.023","url":null,"abstract":"<div><div>Major depressive disorder (MDD) is a multifactorial mental health condition involving genetic, environmental, and neurobiological factors. Conventional antidepressants such as fluoxetine, a selective serotonin reuptake inhibitor, require weeks to exert therapeutic effects, whereas ketamine and esketamine act rapidly via glutamatergic modulation. These drugs may also converge on the inhibition of glycogen synthase kinase 3 beta (<em>GSK3B</em>) as a key mechanism for their antidepressant effects, increasing neuroplasticity, synaptic transmission, and neuronal survival through upregulation of brain-derived neurotrophic factor (<em>BDNF)</em>. Part of the antidepressant effects of ketamine also seems to depend on opioid receptor activation. Despite recent progress, variability in antidepressant response in MDD remains unclear. This work explores, via meta-analysis and network fragility analysis, key molecular mechanisms in MDD, how these drugs exert actions, and highlights potential therapeutic targets for MDD. We performed a network pharmacology approach to unravel the key cellular processes involved in MDD, including altered synaptic plasticity, neurogenesis, apoptosis, and neuroinflammation. Second, we explored the therapeutic role of these treatments on these altered cellular processes. By integrating drug-target data with MDD-associated genes, we identified the opioid receptor mu 1 (<em>OPRM1</em>), epidermal growth factor receptor (<em>EGFR</em>) and <em>GSK3B</em> as key druggable targets. Network analysis further suggested that nuclear factor kappa B (<em>NFKB</em>) may regulate all three, positioning it as a central node linking inflammation, synaptic plasticity, and neuronal metabolism in MDD. We hypothesize that targeted modulation of these genes may optimize the therapeutic efficacy, while <em>NFKB</em> emerges as a promising candidate biomarker for guiding treatment strategies in MDD.</div></div>","PeriodicalId":10715,"journal":{"name":"Computational and structural biotechnology journal","volume":"31 ","pages":"Pages 235-249"},"PeriodicalIF":4.1,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145921291","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-01DOI: 10.1016/j.csbj.2025.12.034
Juan José Oropeza-Valdez , Cristian Padron-Manrique , Jorge E. Arellano-Villavicencio , Aarón Vázquez-Jiménez , Laura E. Hernández-Juárez , Xavier Soberon , María de Lourdes Reyes-Escogido , Rodolfo Guardado-Mendoza , Osbaldo Resendis-Antonio
Prediabetes confers a high risk of progressing to type 2 diabetes mellitus (T2DM). While metformin, a first-line T2DM therapy, improves glycemic control in prediabetes, its effects on the gut microbiota and host metabolic shifts remain poorly understood. Here, we applied a genome-scale community metabolic modeling to build a personalized “digital microbiota” for analyzing the metabolic activity of gut microbes in 106 samples of Mexican prediabetic patients, distributed among patients without treatment and patients treated with metformin over baseline, 6 and 12 months. To contrast microbial metabolic activity across groups and explore how diet modulates it, we simulated computationally the microbial metabolic fluxes under Western, Mediterranean, and traditional Milpa diets across the three groups. As expected, in general terms in silico dietary intervention changes the metabolic responses in the microbiota profiles among the stages, suggesting specific combinations of diets that favor the production of relevant metabolites for wellness, such as amino sugars, short-chain fatty acids, and bile acid exchange fluxes. Furthermore, by selecting two individuals across the entire time as case studies, we provide a proof of concept for in silico personalized diet design. These examples illustrate how the concept of personalized digital microbiota could be leveraged to optimize dietary strategies and potentially improve outcomes in prediabetic patients.
{"title":"Digital modeling of metformin and diet interactions on gut-microbiota metabolism in prediabetic patients","authors":"Juan José Oropeza-Valdez , Cristian Padron-Manrique , Jorge E. Arellano-Villavicencio , Aarón Vázquez-Jiménez , Laura E. Hernández-Juárez , Xavier Soberon , María de Lourdes Reyes-Escogido , Rodolfo Guardado-Mendoza , Osbaldo Resendis-Antonio","doi":"10.1016/j.csbj.2025.12.034","DOIUrl":"10.1016/j.csbj.2025.12.034","url":null,"abstract":"<div><div>Prediabetes confers a high risk of progressing to type 2 diabetes mellitus (T2DM). While metformin, a first-line T2DM therapy, improves glycemic control in prediabetes, its effects on the gut microbiota and host metabolic shifts remain poorly understood. Here, we applied a genome-scale community metabolic modeling to build a personalized “digital microbiota” for analyzing the metabolic activity of gut microbes in 106 samples of Mexican prediabetic patients, distributed among patients without treatment and patients treated with metformin over baseline, 6 and 12 months. To contrast microbial metabolic activity across groups and explore how diet modulates it, we simulated computationally the microbial metabolic fluxes under Western, Mediterranean, and traditional Milpa diets across the three groups. As expected, in general terms <em>in silico</em> dietary intervention changes the metabolic responses in the microbiota profiles among the stages, suggesting specific combinations of diets that favor the production of relevant metabolites for wellness, such as amino sugars, short-chain fatty acids, and bile acid exchange fluxes. Furthermore, by selecting two individuals across the entire time as case studies, we provide a proof of concept for <em>in silico</em> personalized diet design. These examples illustrate how the concept of personalized digital microbiota could be leveraged to optimize dietary strategies and potentially improve outcomes in prediabetic patients.</div></div>","PeriodicalId":10715,"journal":{"name":"Computational and structural biotechnology journal","volume":"31 ","pages":"Pages 250-262"},"PeriodicalIF":4.1,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145921293","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-01DOI: 10.1016/j.csbj.2026.01.009
Rong Xu, Guihua Cao, Liming Hou, Wei Fu, Chenting Bi, Xu Li, Xiaoming Wang
Bisdemethoxycurcumin (BDMC), a natural derivative of curcumin with improved solubility and stability, has shown potential cardioprotective properties. This study investigated the efficacy and underlying mechanisms of BDMC in heart failure with preserved ejection fraction (HFpEF) using both in vivo and in vitro models. The HFpEF mouse model was established using a high-fat diet and L-NAME. BDMC treatment improved cardiac function, attenuated myocardial fibrosis, and exhibited antioxidant effects. Mechanistically, integrated network pharmacology and proteomics identified TGFBR1 as a potential target. BDMC inhibited cardiac fibroblast activation by suppressing TGFBR1 expression and SMAD2/3 phosphorylation. Molecular docking and dynamics simulations confirmed stable binding between BDMC and TGFBR1. These findings demonstrate that BDMC mitigates myocardial fibrosis in HFpEF, primarily by competitively inhibiting the binding of TGF-β and TGFBR1, achieving the effect of inhibiting cardiac fibroblast activation.
{"title":"Bisdemethoxycurcumin attenuates myocardial fibrosis in heart failure with preserved ejection fraction by targeting TGFBR1 and oxidative stress","authors":"Rong Xu, Guihua Cao, Liming Hou, Wei Fu, Chenting Bi, Xu Li, Xiaoming Wang","doi":"10.1016/j.csbj.2026.01.009","DOIUrl":"10.1016/j.csbj.2026.01.009","url":null,"abstract":"<div><div>Bisdemethoxycurcumin (BDMC), a natural derivative of curcumin with improved solubility and stability, has shown potential cardioprotective properties. This study investigated the efficacy and underlying mechanisms of BDMC in heart failure with preserved ejection fraction (HFpEF) using both <em>in vivo</em> and <em>in vitro</em> models. The HFpEF mouse model was established using a high-fat diet and <span>L</span>-NAME. BDMC treatment improved cardiac function, attenuated myocardial fibrosis, and exhibited antioxidant effects. Mechanistically, integrated network pharmacology and proteomics identified TGFBR1 as a potentia<u>l</u> target. BDMC inhibited cardiac fibroblast activation by suppressing TGFBR1 expression and SMAD2/3 phosphorylation. Molecular docking and dynamics simulations confirmed stable binding between BDMC and TGFBR1. These findings demonstrate that BDMC mitigates myocardial fibrosis in HFpEF, primarily by competitively inhibiting the binding of TGF-β and TGFBR1, achieving the effect of inhibiting cardiac fibroblast activation.</div></div>","PeriodicalId":10715,"journal":{"name":"Computational and structural biotechnology journal","volume":"31 ","pages":"Pages 422-435"},"PeriodicalIF":4.1,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145973254","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-01DOI: 10.1016/j.csbj.2026.01.003
Kewei Song , Tao Zhang , Xin Xie , DeHua Liao , Linlin Wu , Pei Jiang
The aim of this study is to investigate the gene biomarkers and metabolites for esophageal squamous cell carcinoma (ESCC) or cisplatin (DDP)-resistance ESCC through integrated analysis of transcriptome and metabolome. A total of 6130 differentially expressed genes (DEGs) and 326 differentially expressed metabolites (DEMs) were identified in KYSE30 compared to HEEC, while compared to KYSE30, there were totally 1179 DEGs and 224 DEMs in KYSE30/DDP. Profile #6 depicted the mRNA characteristics of KYSE30 obviously. Genes in profile #6 were mainly involved in platinum drug resistance, ferroptosis, and glutathione metabolism. In addition, the associated TCGA dataset identified APOBEC3B as a critical gene involved in inhibiting ferroptosis by activating PD-L1 to suppress CD8+T cells. In vitro experiments demonstrated that knockdown of APOBEC3B enhanced ferroptosis and inhibited the glutathione metabolism signaling pathway in KYSE30/DDP. Moreover, in vivo experiments further confirmed that knockdown of APOBEC3B suppressed PD-L1, thereby activating CD8+T cells and promoting ferroptosis. These findings indicate the critical role of ferroptosis and glutathione metabolism in the development and progression of ESCC. Meanwhile, APOBEC3B may serve as a promising therapeutic target for cisplatin-resistant ESCC cells.
{"title":"Integrated metabolome and transcriptome analysis reveals ferroptosis involvement in cisplatin resistance of esophageal squamous cancer cell","authors":"Kewei Song , Tao Zhang , Xin Xie , DeHua Liao , Linlin Wu , Pei Jiang","doi":"10.1016/j.csbj.2026.01.003","DOIUrl":"10.1016/j.csbj.2026.01.003","url":null,"abstract":"<div><div>The aim of this study is to investigate the gene biomarkers and metabolites for esophageal squamous cell carcinoma (ESCC) or cisplatin (DDP)-resistance ESCC through integrated analysis of transcriptome and metabolome. A total of 6130 differentially expressed genes (DEGs) and 326 differentially expressed metabolites (DEMs) were identified in KYSE30 compared to HEEC, while compared to KYSE30, there were totally 1179 DEGs and 224 DEMs in KYSE30/DDP. Profile #6 depicted the mRNA characteristics of KYSE30 obviously. Genes in profile #6 were mainly involved in platinum drug resistance, ferroptosis, and glutathione metabolism. In addition, the associated TCGA dataset identified APOBEC3B as a critical gene involved in inhibiting ferroptosis by activating PD-L1 to suppress CD8<sup>+</sup>T cells. In vitro experiments demonstrated that knockdown of APOBEC3B enhanced ferroptosis and inhibited the glutathione metabolism signaling pathway in KYSE30/DDP. Moreover, in vivo experiments further confirmed that knockdown of APOBEC3B suppressed PD-L1, thereby activating CD8<sup>+</sup>T cells and promoting ferroptosis. These findings indicate the critical role of ferroptosis and glutathione metabolism in the development and progression of ESCC. Meanwhile, APOBEC3B may serve as a promising therapeutic target for cisplatin-resistant ESCC cells.</div></div>","PeriodicalId":10715,"journal":{"name":"Computational and structural biotechnology journal","volume":"31 ","pages":"Pages 397-411"},"PeriodicalIF":4.1,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145973106","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-01DOI: 10.1016/j.csbj.2026.01.008
Ting Kou , Jue Wang , Jing Zhou , Yang Cheng , Lei Xu , Jinpeng Xie , Ying Chen , Ting Zhang , Shuaifei Zhao , Yuxiu Ying , Xiaoshuang Xin , Xu Xu , Dandan Lu , Xiangyu Hu , Chenyu Qiu , Jun Wang , Gu Cheng , Siyun Lei , Qiqi Lyu , Yihuai Pan , Tong Cao
Mesenchymal stem cells (MSCs) are widely applied in regenerative medicine, but conventional osteogenic induction assays are time-consuming and rely on destructive endpoint measurements. Here, we propose OsteoNet, a deep learning framework that predicts osteogenic differentiation from bright-field images and generates an Osteogenic Score (OsScore) reflecting differentiation dynamics. The predictive performance of OsteoNet was evaluated across multiple time points using an independent test set, achieving an AUC of 0.94 on day 0 and 0.98 on day 5, demonstrating robust early-stage detection capability. The OsScore increased progressively with induction time and showed strong positive correlations with both early and late osteogenic markers, including RUNX2, OCN, and OSX, at the RNA and protein levels. Morphological analysis of immunofluorescence images further confirmed significant increases in cell size during early differentiation, supporting the model’s sensitivity to subtle morphological cues. Collectively, OsteoNet enables non-invasive, quantitative, and early monitoring of osteogenic differentiation in hAMSCs, offering a powerful tool to accelerate research and reduce reliance on destructive endpoint assays.
{"title":"OsteoNet: A deep learning framework linking cellular morphology to molecular markers for quantifying osteogenic differentiation","authors":"Ting Kou , Jue Wang , Jing Zhou , Yang Cheng , Lei Xu , Jinpeng Xie , Ying Chen , Ting Zhang , Shuaifei Zhao , Yuxiu Ying , Xiaoshuang Xin , Xu Xu , Dandan Lu , Xiangyu Hu , Chenyu Qiu , Jun Wang , Gu Cheng , Siyun Lei , Qiqi Lyu , Yihuai Pan , Tong Cao","doi":"10.1016/j.csbj.2026.01.008","DOIUrl":"10.1016/j.csbj.2026.01.008","url":null,"abstract":"<div><div>Mesenchymal stem cells (MSCs) are widely applied in regenerative medicine, but conventional osteogenic induction assays are time-consuming and rely on destructive endpoint measurements. Here, we propose OsteoNet, a deep learning framework that predicts osteogenic differentiation from bright-field images and generates an Osteogenic Score (OsScore) reflecting differentiation dynamics. The predictive performance of OsteoNet was evaluated across multiple time points using an independent test set, achieving an AUC of 0.94 on day 0 and 0.98 on day 5, demonstrating robust early-stage detection capability. The OsScore increased progressively with induction time and showed strong positive correlations with both early and late osteogenic markers, including RUNX2, OCN, and OSX, at the RNA and protein levels. Morphological analysis of immunofluorescence images further confirmed significant increases in cell size during early differentiation, supporting the model’s sensitivity to subtle morphological cues. Collectively, OsteoNet enables non-invasive, quantitative, and early monitoring of osteogenic differentiation in hAMSCs, offering a powerful tool to accelerate research and reduce reliance on destructive endpoint assays.</div></div>","PeriodicalId":10715,"journal":{"name":"Computational and structural biotechnology journal","volume":"31 ","pages":"Pages 436-447"},"PeriodicalIF":4.1,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145973107","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-01DOI: 10.1016/j.csbj.2025.12.024
Amal Bouzid , Ayesha M. Yusuf , Mira Mousa , Thenmozhi Venkatachalam , Guan Tay , Maimunah Uddin , Nawal Alkaabi , Maha Saber Ayad , Habiba Alsafar , Rifat Hamoudi
Background & objective
Emerging viral infections can initiate a global pandemic with high mortality. Understanding the immunopathogenesis of these viruses is critical to developing effective strategies for managing/preventing such outbreaks.
Methods
Transcriptomics analysis was performed in a UAE cohort with respective COVID-19 severities. The findings were correlated with published studies of COVID-19 severity/progression GWAS, and transcriptomics data of patients infected with different respiratory viruses.
Results
The transcriptional profiling distinguished significantly between the infected COVID-19 groups and identified the interferon-induced protein, GBP2, as a common significantly up-regulated gene among the different COVID-19 infection severities (nominal p = 0.0019). Key inflammatory pathways were enriched in the higher-severity groups, including Interleukin-1 family signaling. A remarkable immune signature resulted in a trend of cytokine expression changes between all severities, including CCL19, CCL21, IL-19, IL-20, IL-36RN, and members of the IFNA family. The deconvolution of immune cells showed a trend of an uncontrolled pro-inflammatory state and poor immune function in higher disease severities. A systematic analysis of the transcriptomic and GWAS findings identified common signature genes between COVID-19 infection severities including ALCAM, DKK3, EFNA5, FN1, GABRA5, LPAR1, METTL8, MTHFD1L, SPOCK1, TPM4, VTI1A, and WWC2. Differential regulation in potential genes associated with the Interferon signaling pathway including HERC5, IFFI44L, IFI6, RSAD2 and SP100 was identified as a common feature in transcriptomes of patients afflicted with different virulent respiratory viruses.
Conclusion
Our findings highlight a direction where changes in immune response and specific biomarker panels could be considered as a strategy for the prediction/prevention of new emerging respiratory virus outbreaks.
{"title":"COVID-19 immunopathological features for the prediction and prevention of future emerging respiratory viral infections","authors":"Amal Bouzid , Ayesha M. Yusuf , Mira Mousa , Thenmozhi Venkatachalam , Guan Tay , Maimunah Uddin , Nawal Alkaabi , Maha Saber Ayad , Habiba Alsafar , Rifat Hamoudi","doi":"10.1016/j.csbj.2025.12.024","DOIUrl":"10.1016/j.csbj.2025.12.024","url":null,"abstract":"<div><h3>Background & objective</h3><div>Emerging viral infections can initiate a global pandemic with high mortality. Understanding the immunopathogenesis of these viruses is critical to developing effective strategies for managing/preventing such outbreaks.</div></div><div><h3>Methods</h3><div>Transcriptomics analysis was performed in a UAE cohort with respective COVID-19 severities. The findings were correlated with published studies of COVID-19 severity/progression GWAS, and transcriptomics data of patients infected with different respiratory viruses.</div></div><div><h3>Results</h3><div>The transcriptional profiling distinguished significantly between the infected COVID-19 groups and identified the interferon-induced protein, <em>GBP2,</em> as a common significantly up-regulated gene among the different COVID-19 infection severities (nominal <em>p</em> = 0.0019). Key inflammatory pathways were enriched in the higher-severity groups, including Interleukin-1 family signaling. A remarkable immune signature resulted in a trend of cytokine expression changes between all severities, including <em>CCL19, CCL21, IL-19, IL-20, IL-36RN,</em> and members of the IFNA family. The deconvolution of immune cells showed a trend of an uncontrolled pro-inflammatory state and poor immune function in higher disease severities. A systematic analysis of the transcriptomic and GWAS findings identified common signature genes between COVID-19 infection severities including <em>ALCAM, DKK3, EFNA5, FN1, GABRA5, LPAR1, METTL8, MTHFD1L, SPOCK1, TPM4, VTI1A,</em> and <em>WWC2.</em> Differential regulation in potential genes associated with the Interferon signaling pathway including <em>HERC5, IFFI44L, IFI6, RSAD2</em> and <em>SP100</em> was identified as a common feature in transcriptomes of patients afflicted with different virulent respiratory viruses.</div></div><div><h3>Conclusion</h3><div>Our findings highlight a direction where changes in immune response and specific biomarker panels could be considered as a strategy for the prediction/prevention of new emerging respiratory virus outbreaks.</div></div>","PeriodicalId":10715,"journal":{"name":"Computational and structural biotechnology journal","volume":"32 ","pages":"Pages 1-15"},"PeriodicalIF":4.1,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145883391","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-01DOI: 10.1016/j.csbj.2025.11.053
Darren Fang , Dan Ruan
Background
Wearable devices enable continuous acquisition of physiological and behavioral signals with broad utility in precision and population health. However, real-world datasets often have privacy constraints, irregular sampling, and activity-dependent recording, complicating data sharing and modeling. We present a synthesis framework that preserves statistical realism by matching marginal distributions and inter-record distances.
Method
We propose a hierarchical heated Markov model (HHMM) that captures conditional dependencies and time-varying behavioral patterns. Tier 1 generates minute-level activity types using hour-specific priors and a heating mechanism to achieve a target ergodic distribution. Tier 2 samples heart rate (via Metropolis–Hastings conditioned on activity) and activity duration from empirical conditional distributions. Tier 3 synthesizes additional variables (e.g., sleep duration, calories) conditioned on Tiers 1–2, with deterministic rules for activity codes and floors climbed. Activity-conditioned Poisson subsampling emulates device-driven irregular timestamps. We benchmark HHMM against CTGAN and TVAE using a synthetic IEEE BHI dataset and validate on a real Fitbit dataset. Fidelity is assessed by comparing distributions of pairwise inter-record distances—within synthetic vs. within reference cohorts—via Wasserstein distances for Kolmogorov–Smirnov (KS), Jensen–Shannon (JSD), and distance-correlation (DC) metrics.
Results
On the BHI dataset, mean WD(KS)/WD(JSD)/WD(DC) were 0.125/0.242/0.327 for HHMM, 0.130/0.245/0.257 for CTGAN, and 0.129/0.246/0.249 for TVAE. On the Fitbit dataset, values were 0.295/0.489/0.233 (HHMM), 0.293/0.488/0.209 (CTGAN), and 0.293/0.588/0.160 (TVAE).
Discussion
HHMM offers task-specific gains on synthetic benchmarks. Real-world results highlight a need for domain adaptation. The method is computationally efficient and privacy-preserving, supporting scalable synthetic data generation for wearable health research.
{"title":"Hierarchical heated markov modeling for synthesizing activity data from wearable devices","authors":"Darren Fang , Dan Ruan","doi":"10.1016/j.csbj.2025.11.053","DOIUrl":"10.1016/j.csbj.2025.11.053","url":null,"abstract":"<div><h3>Background</h3><div>Wearable devices enable continuous acquisition of physiological and behavioral signals with broad utility in precision and population health. However, real-world datasets often have privacy constraints, irregular sampling, and activity-dependent recording, complicating data sharing and modeling. We present a synthesis framework that preserves statistical realism by matching marginal distributions and inter-record distances.</div></div><div><h3>Method</h3><div>We propose a hierarchical heated Markov model (HHMM) that captures conditional dependencies and time-varying behavioral patterns. Tier 1 generates minute-level activity types using hour-specific priors and a heating mechanism to achieve a target ergodic distribution. Tier 2 samples heart rate (via Metropolis–Hastings conditioned on activity) and activity duration from empirical conditional distributions. Tier 3 synthesizes additional variables (e.g., sleep duration, calories) conditioned on Tiers 1–2, with deterministic rules for activity codes and floors climbed. Activity-conditioned Poisson subsampling emulates device-driven irregular timestamps. We benchmark HHMM against CTGAN and TVAE using a synthetic IEEE BHI dataset and validate on a real Fitbit dataset. Fidelity is assessed by comparing distributions of pairwise inter-record distances—within synthetic vs. within reference cohorts—via Wasserstein distances for Kolmogorov–Smirnov (KS), Jensen–Shannon (JSD), and distance-correlation (DC) metrics.</div></div><div><h3>Results</h3><div>On the BHI dataset, mean WD(KS)/WD(JSD)/WD(DC) were 0.125/0.242/0.327 for HHMM, 0.130/0.245/0.257 for CTGAN, and 0.129/0.246/0.249 for TVAE. On the Fitbit dataset, values were 0.295/0.489/0.233 (HHMM), 0.293/0.488/0.209 (CTGAN), and 0.293/0.588/0.160 (TVAE).</div></div><div><h3>Discussion</h3><div>HHMM offers task-specific gains on synthetic benchmarks. Real-world results highlight a need for domain adaptation. The method is computationally efficient and privacy-preserving, supporting scalable synthetic data generation for wearable health research.</div></div>","PeriodicalId":10715,"journal":{"name":"Computational and structural biotechnology journal","volume":"32 ","pages":"Pages 16-23"},"PeriodicalIF":4.1,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145923438","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-01DOI: 10.1016/j.csbj.2025.12.021
Hanife Sahin , Shariful Islam , A.Hyeon Lee , Irene Ponzo , Kilian Winiger , Andreas Reichl , Thomas Carell , Pascal Giehr
5-Hydroxymethyl-2′-deoxycytidine (5hmdC) is an important epigenetic marker involved in gene regulation and DNA demethylation. It has potential use as a biomarker for cancer and other diseases due to its significant depletion in various cancers and disease models. This research aimed to develop a reliable and efficient method for generating 5hmdC-containing DNA, addressing limitations in existing techniques. We created a Tet3 stalling mutant that converts 5-methyl-2′-deoxycytidine (5mdC) into a mixture of 5hmdC and 5-formyl-2′-deoxycytidine (5fdC), followed by a reduction step to convert 5fdC to 5hmdC, ensuring a pure 5hmdC state within the CpG context. This method can convert any PCR product, synthetic oligos, and entire genomes into 5hmdC-modified DNA. The principal results demonstrate high specificity and efficiency, providing a robust tool for epigenetic research, cancer diagnostics, and protein binding assays. Additionally, our technique offers 5hmdC-DNA for functional studies and as standards for diagnostic assays.
{"title":"Biochemical engineering of 5hmdC-DNA using a Tet3 double-mutant","authors":"Hanife Sahin , Shariful Islam , A.Hyeon Lee , Irene Ponzo , Kilian Winiger , Andreas Reichl , Thomas Carell , Pascal Giehr","doi":"10.1016/j.csbj.2025.12.021","DOIUrl":"10.1016/j.csbj.2025.12.021","url":null,"abstract":"<div><div>5-Hydroxymethyl-2′-deoxycytidine (5hmdC) is an important epigenetic marker involved in gene regulation and DNA demethylation. It has potential use as a biomarker for cancer and other diseases due to its significant depletion in various cancers and disease models. This research aimed to develop a reliable and efficient method for generating 5hmdC-containing DNA, addressing limitations in existing techniques. We created a Tet3 stalling mutant that converts 5-methyl-2′-deoxycytidine (5mdC) into a mixture of 5hmdC and 5-formyl-2′-deoxycytidine (5fdC), followed by a reduction step to convert 5fdC to 5hmdC, ensuring a pure 5hmdC state within the CpG context. This method can convert any PCR product, synthetic oligos, and entire genomes into 5hmdC-modified DNA. The principal results demonstrate high specificity and efficiency, providing a robust tool for epigenetic research, cancer diagnostics, and protein binding assays. Additionally, our technique offers 5hmdC-DNA for functional studies and as standards for diagnostic assays.</div></div>","PeriodicalId":10715,"journal":{"name":"Computational and structural biotechnology journal","volume":"31 ","pages":"Pages 389-396"},"PeriodicalIF":4.1,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145973105","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2026-01-01DOI: 10.1016/j.csbj.2025.12.035
Minyoung So , Soo Jung Park , Dongin Kim , Seokjin Han , Hee Jung Koo , Taeyong Kim , Min-Gi Shin , Eun Jeong Lee
The development of disease-modifying therapies (DMTs) for Parkinson’s disease (PD) remains a critical unmet need. Despite extensive research efforts, no therapy capable of slowing or halting PD progression has been approved. Here, we apply a knowledge graph–based artificial intelligence (AI) framework, combined with subgraph-level enrichment–based re-prioritization, to identify novel PD-modifying targets without requiring disease-specific training or additional experimental datasets. Using model-derived PD association scores, we obtained 2527 predicted targets. To evaluate their connectivity to an expert-curated set of PD-associated genes, we performed subgraph-level over-representation analysis and identified 74 targets whose local subgraphs were significantly enriched for PD-relevant context. After applying novelty filters, five candidates remained, among which tripeptidyl peptidase 1 (TPP1) emerged as a compelling PD DMT target. The predicted association among PD, α-synuclein, and TPP1 within the subgraph was supported by differential expression analyses of publicly available RNA-seq datasets and validated experimentally in a human cell–based α-synuclein aggregation model. TPP1 expression was elevated in neuromelanin-positive dopaminergic neurons in late-stage PD, and its knockdown increased α-synuclein aggregation, suggesting a protective role in α-synuclein homeostasis. Structural modeling of AlphaFold-Multimer further revealed a substrate-like interface between α-synuclein and the TPP1 catalytic triad, consistent with a potential proteolytic mechanism of α-synuclein clearance. Together, these findings identify TPP1 as a previously underappreciated and mechanistically plausible PD DMT target and demonstrate how static knowledge graphs can be transformed into interpretable, disease-focused target discovery systems. By integrating explainable subgraph structures with enrichment-based re-prioritization, this framework provides a generalizable strategy for therapeutic target identification across indications.
{"title":"Systematic discovery of disease-modifying targets by prediction from knowledge graph-based AI model and experimental validation: Parkinson’s disease case","authors":"Minyoung So , Soo Jung Park , Dongin Kim , Seokjin Han , Hee Jung Koo , Taeyong Kim , Min-Gi Shin , Eun Jeong Lee","doi":"10.1016/j.csbj.2025.12.035","DOIUrl":"10.1016/j.csbj.2025.12.035","url":null,"abstract":"<div><div>The development of disease-modifying therapies (DMTs) for Parkinson’s disease (PD) remains a critical unmet need. Despite extensive research efforts, no therapy capable of slowing or halting PD progression has been approved. Here, we apply a knowledge graph–based artificial intelligence (AI) framework, combined with subgraph-level enrichment–based re-prioritization, to identify novel PD-modifying targets without requiring disease-specific training or additional experimental datasets. Using model-derived PD association scores, we obtained 2527 predicted targets. To evaluate their connectivity to an expert-curated set of PD-associated genes, we performed subgraph-level over-representation analysis and identified 74 targets whose local subgraphs were significantly enriched for PD-relevant context. After applying novelty filters, five candidates remained, among which tripeptidyl peptidase 1 (TPP1) emerged as a compelling PD DMT target. The predicted association among PD, α-synuclein, and TPP1 within the subgraph was supported by differential expression analyses of publicly available RNA-seq datasets and validated experimentally in a human cell–based α-synuclein aggregation model. <em>TPP1</em> expression was elevated in neuromelanin-positive dopaminergic neurons in late-stage PD, and its knockdown increased α-synuclein aggregation, suggesting a protective role in α-synuclein homeostasis. Structural modeling of AlphaFold-Multimer further revealed a substrate-like interface between α-synuclein and the TPP1 catalytic triad, consistent with a potential proteolytic mechanism of α-synuclein clearance. Together, these findings identify TPP1 as a previously underappreciated and mechanistically plausible PD DMT target and demonstrate how static knowledge graphs can be transformed into interpretable, disease-focused target discovery systems. By integrating explainable subgraph structures with enrichment-based re-prioritization, this framework provides a generalizable strategy for therapeutic target identification across indications.</div></div>","PeriodicalId":10715,"journal":{"name":"Computational and structural biotechnology journal","volume":"31 ","pages":"Pages 289-300"},"PeriodicalIF":4.1,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145921292","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}