Pub Date : 2025-10-24eCollection Date: 2025-01-01DOI: 10.7150/ijbs.124144
Zimo Jia, Jiajin Wu, Jiyue Zhang, Peixian Zheng, Haoxuan Zhang, Yiqin Lin, Tao Pan, Meng Wu, Yuqin Song
Chimeric antigen receptor (CAR)-T cell therapy represents a breakthrough in cancer immunotherapy, demonstrating impressive clinical outcomes, particularly for hematologic malignancies. However, its broader therapeutic application, especially against solid tumors, remains limited. Key challenges include T cell exhaustion, limited persistence, cytokine-mediated toxicities, and logistical hurdles associated with manufacturing autologous products. Emerging gene editing technologies, such as CRISPR/Cas systems, base editing, and prime editing, offer novel approaches to optimize CAR-T cells, aiming to enhance efficacy while managing toxicity and improving accessibility. This review comprehensively examines the current landscape of these gene editing tools in CAR-T cell therapy, highlighting the latest advancements, persisting challenges, and future directions. Leveraging gene editing holds the potential to transform CAR-T therapy into a more potent, safer, and broadly applicable modality for cancer and beyond.
{"title":"Precision Reprogramming in CAR-T Cell Therapy: Innovations, Challenges, and Future Directions of Advanced Gene Editing.","authors":"Zimo Jia, Jiajin Wu, Jiyue Zhang, Peixian Zheng, Haoxuan Zhang, Yiqin Lin, Tao Pan, Meng Wu, Yuqin Song","doi":"10.7150/ijbs.124144","DOIUrl":"10.7150/ijbs.124144","url":null,"abstract":"<p><p>Chimeric antigen receptor (CAR)-T cell therapy represents a breakthrough in cancer immunotherapy, demonstrating impressive clinical outcomes, particularly for hematologic malignancies. However, its broader therapeutic application, especially against solid tumors, remains limited. Key challenges include T cell exhaustion, limited persistence, cytokine-mediated toxicities, and logistical hurdles associated with manufacturing autologous products. Emerging gene editing technologies, such as CRISPR/Cas systems, base editing, and prime editing, offer novel approaches to optimize CAR-T cells, aiming to enhance efficacy while managing toxicity and improving accessibility. This review comprehensively examines the current landscape of these gene editing tools in CAR-T cell therapy, highlighting the latest advancements, persisting challenges, and future directions. Leveraging gene editing holds the potential to transform CAR-T therapy into a more potent, safer, and broadly applicable modality for cancer and beyond.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":"21 15","pages":"6884-6906"},"PeriodicalIF":10.0,"publicationDate":"2025-10-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12631245/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145587261","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 : 2025-10-24eCollection Date: 2025-01-01DOI: 10.7150/ijbs.119301
Diego Liviu Boaru, Diego De Leon-Oliva, Patricia De Castro-Martinez, Cielo Garcia-Montero, Oscar Fraile-Martinez, Beatriz García-González, Isabel Pérez-González, Majd N Michael Alhaddadin, Silvestra Barrena-Blázquez, Laura Lopez-Gonzalez, Basilio de la Torre, Leonel Pekarek, Miguel A Saez, Laura Ríos-Espinosa, Tatiana Pekarek, Roberto Fernández-Baillo Gallego de la Sacristana, Mauricio Hernández-Fernández, Carlos Casanova, Ana Castel-Oñate, Natalio Garcia-Honduvilla, Julia Buján, Raul Diaz-Pedrero, Melchor Alvarez-Mon, Miguel A Ortega
This review underscores the dynamic role of the extracellular matrix (ECM) in regulating cellular behavior and maintaining tissue homeostasis, highlighting its pivotal involvement in aging, calcification, and cancer diseases. In healthy tissues, controlled ECM remodeling provides essential biochemical and mechanical cues, but dysregulation (driven by chronic inflammation, cellular senescence, and altered intercellular communication) leads to fibrosis, calcification, and the creation of a pro-tumorigenic microenvironment. Senescent cells contribute to these changes through senescence-associated secretory phenotype (SASP), which reinforces inflammation and matrix degradation, while extracellular vesicles (EVs) mediate intercellular signaling and further modulate ECM structure and function. In cancer, ECM remodeling not only facilitates tumor progression and metastasis by forming physical and biochemical barriers but also hinders the efficacy of conventional and immunotherapeutic interventions. Similarly, in cardiovascular diseases, aberrant ECM remodeling exacerbates tissue damage and impairs regenerative processes. Emerging therapeutic strategies aim to restore ECM homeostasis through targeted interventions, including ECM-normalizing agents, EV-based therapies, and stem cell approaches that modulate matrix composition to improve tissue repair. By elucidating the complex interplay between ECM dysfunction, cellular senescence, and chronic inflammation, this review highlights promising avenues for developing personalized treatments that address the underlying causes of age-related and tumorigenic pathologies, ultimately, the way to improved clinical outcomes.
{"title":"Extracellular matrix dysregulation in aging, calcification, and cancer diseases: insights into cellular senescence, inflammation, and novel therapeutic strategies.","authors":"Diego Liviu Boaru, Diego De Leon-Oliva, Patricia De Castro-Martinez, Cielo Garcia-Montero, Oscar Fraile-Martinez, Beatriz García-González, Isabel Pérez-González, Majd N Michael Alhaddadin, Silvestra Barrena-Blázquez, Laura Lopez-Gonzalez, Basilio de la Torre, Leonel Pekarek, Miguel A Saez, Laura Ríos-Espinosa, Tatiana Pekarek, Roberto Fernández-Baillo Gallego de la Sacristana, Mauricio Hernández-Fernández, Carlos Casanova, Ana Castel-Oñate, Natalio Garcia-Honduvilla, Julia Buján, Raul Diaz-Pedrero, Melchor Alvarez-Mon, Miguel A Ortega","doi":"10.7150/ijbs.119301","DOIUrl":"10.7150/ijbs.119301","url":null,"abstract":"<p><p>This review underscores the dynamic role of the extracellular matrix (ECM) in regulating cellular behavior and maintaining tissue homeostasis, highlighting its pivotal involvement in aging, calcification, and cancer diseases. In healthy tissues, controlled ECM remodeling provides essential biochemical and mechanical cues, but dysregulation (driven by chronic inflammation, cellular senescence, and altered intercellular communication) leads to fibrosis, calcification, and the creation of a pro-tumorigenic microenvironment. Senescent cells contribute to these changes through senescence-associated secretory phenotype (SASP), which reinforces inflammation and matrix degradation, while extracellular vesicles (EVs) mediate intercellular signaling and further modulate ECM structure and function. In cancer, ECM remodeling not only facilitates tumor progression and metastasis by forming physical and biochemical barriers but also hinders the efficacy of conventional and immunotherapeutic interventions. Similarly, in cardiovascular diseases, aberrant ECM remodeling exacerbates tissue damage and impairs regenerative processes. Emerging therapeutic strategies aim to restore ECM homeostasis through targeted interventions, including ECM-normalizing agents, EV-based therapies, and stem cell approaches that modulate matrix composition to improve tissue repair. By elucidating the complex interplay between ECM dysfunction, cellular senescence, and chronic inflammation, this review highlights promising avenues for developing personalized treatments that address the underlying causes of age-related and tumorigenic pathologies, ultimately, the way to improved clinical outcomes.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":"21 15","pages":"6808-6881"},"PeriodicalIF":10.0,"publicationDate":"2025-10-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12631191/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145587274","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 : 2025-10-24eCollection Date: 2025-01-01DOI: 10.7150/ijbs.102132
Mouad Edderkaoui, Omer H M Elmadbouh, Adrian Lim, Yan Ou, Dina Hauptschein, Ankita Guha, Abdo Darwish, Vinicius F Calsavara, Ramachandran Murali, Neil Bhowmick, Arsen Osipov, Angela J Mathison, Raul Urrutia, Qiang Wang, Stephen J Pandol
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest forms of human malignancy, and there is an urgency to develop more effective therapy. We previously showed that Metavert, a dual inhibitor of glycogen synthase kinase 3-beta (GSK-3β) and histone deacetylases (HDACs) prevents pancreatic ductal adenocarcinoma (PDAC) metastasis. In this study, we investigated the mechanisms that mediate metastasis and the roles of GSK-3β, HDACs, and Yes-associated protein (YAP) in this process. We found that HDAC4 and YAP are highly expressed in PDAC from patients with rapid disease progression and metastasis compared to those with prolonged recurrence-free survival. Pan-HDAC inhibition decreases metastasis in the splenic PDAC metastatic mouse model. Inhibition of HDAC4 reduces migration of cancer cells and decreases the mRNA and protein levels of transcription factor MYB Proto-Oncogene Like 1 (MybL1) and YAP. Mechanistic studies show that HDAC4 regulates transcription of YAP through up-regulating MybL1 expression. Comparable results were observed in colon and prostate cancers. ATAC-seq studies show that inhibition of HDAC4 causes chromatin remodeling in the YAP promoter region and reduces accessibility to the binding sites of multiple transcription factors, including those of MybL1. Pharmacological or molecular inhibition of YAP significantly decreases PDAC metastasis in vivo. Imaging Mass Cytometry (IMC) reveals no significant changes in immune cells, but a notable shift in the distribution patterns of cancer-associated hepatic stellate cells in the metastatic niche, when YAP is ablated in the cancer cells. The results demonstrate a novel metastasis-driving cell signaling pathway mediated by the functional interaction between HDAC4 and MybL1, which regulates YAP expression and metastasis.
胰腺导管腺癌(PDAC)是人类最致命的恶性肿瘤之一,迫切需要开发更有效的治疗方法。我们之前的研究表明,糖原合成酶激酶3- β (GSK-3β)和组蛋白去乙酰化酶(hdac)的双重抑制剂Metavert可以阻止胰腺导管腺癌(PDAC)的转移。在这项研究中,我们探讨了介导转移的机制以及GSK-3β、hdac和Yes-associated protein (YAP)在这一过程中的作用。我们发现,与无复发生存期较长的患者相比,快速疾病进展和转移患者的PDAC中HDAC4和YAP高表达。抑制泛hdac可减少脾PDAC转移小鼠模型的转移。抑制HDAC4可减少癌细胞的迁移,降低转录因子MYB原癌基因样1 (MybL1)和YAP的mRNA和蛋白水平。机制研究表明,HDAC4通过上调MybL1表达来调控YAP的转录。在结肠癌和前列腺癌中也观察到类似的结果。ATAC-seq研究表明,抑制HDAC4会导致YAP启动子区域的染色质重塑,并降低多种转录因子结合位点的可及性,包括MybL1的结合位点。药理或分子抑制YAP可显著降低PDAC体内转移。成像细胞计数(IMC)显示免疫细胞无明显变化,但当YAP在癌细胞中消融时,转移生态位中与癌症相关的肝星状细胞的分布模式发生了显著变化。结果表明,HDAC4和MybL1之间的功能相互作用介导了一种新的转移驱动细胞信号通路,该信号通路调节YAP的表达和转移。
{"title":"HDAC4/MybL1/YAP novel signaling axis is required for pancreatic cancer metastasis to the liver.","authors":"Mouad Edderkaoui, Omer H M Elmadbouh, Adrian Lim, Yan Ou, Dina Hauptschein, Ankita Guha, Abdo Darwish, Vinicius F Calsavara, Ramachandran Murali, Neil Bhowmick, Arsen Osipov, Angela J Mathison, Raul Urrutia, Qiang Wang, Stephen J Pandol","doi":"10.7150/ijbs.102132","DOIUrl":"10.7150/ijbs.102132","url":null,"abstract":"<p><p>Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest forms of human malignancy, and there is an urgency to develop more effective therapy. We previously showed that Metavert, a dual inhibitor of glycogen synthase kinase 3-beta (GSK-3β) and histone deacetylases (HDACs) prevents pancreatic ductal adenocarcinoma (PDAC) metastasis. In this study, we investigated the mechanisms that mediate metastasis and the roles of GSK-3β, HDACs, and Yes-associated protein (YAP) in this process. We found that HDAC4 and YAP are highly expressed in PDAC from patients with rapid disease progression and metastasis compared to those with prolonged recurrence-free survival. Pan-HDAC inhibition decreases metastasis in the splenic PDAC metastatic mouse model. Inhibition of HDAC4 reduces migration of cancer cells and decreases the mRNA and protein levels of transcription factor MYB Proto-Oncogene Like 1 (MybL1) and YAP. Mechanistic studies show that HDAC4 regulates transcription of YAP through up-regulating MybL1 expression. Comparable results were observed in colon and prostate cancers. ATAC-seq studies show that inhibition of HDAC4 causes chromatin remodeling in the YAP promoter region and reduces accessibility to the binding sites of multiple transcription factors, including those of MybL1. Pharmacological or molecular inhibition of YAP significantly decreases PDAC metastasis in vivo. Imaging Mass Cytometry (IMC) reveals no significant changes in immune cells, but a notable shift in the distribution patterns of cancer-associated hepatic stellate cells in the metastatic niche, when YAP is ablated in the cancer cells. The results demonstrate a novel metastasis-driving cell signaling pathway mediated by the functional interaction between HDAC4 and MybL1, which regulates YAP expression and metastasis.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":"21 15","pages":"6907-6925"},"PeriodicalIF":10.0,"publicationDate":"2025-10-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12631223/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145587183","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 : 2025-10-20eCollection Date: 2025-01-01DOI: 10.7150/ijbs.116383
Tinghui Jiang, Xin Liu, Hao Liu, Kailong Du, Yitao Wang, Hui Fan, Ying Zhang, Lin Cui, Hewei Zhang, Chundong Zhang, Yong Zhu, Zhongyu Liu, Youquan Bu, Yunlong Lei
Colorectal cancer (CRC) is one of the most common malignant cancers worldwide and its poor prognosis is mainly caused by metastasis. Although extensive studies, the potential molecular mechanisms of CRC metastasis are not fully understood. In the present study, we found that ETV4 was remarkably upregulated in CRC and its overexpression correlated with lymph node metastasis. ETV4 could significantly promote the growth, epithelial-mesenchymal transition (EMT) and metastasis of CRC cells in vitro and in vivo. Mechanistic investigations found that LOXL2 was a novel transcriptional target and a direct interacting partner of ETV4 and was vital to ETV4-induced CRC malignant phenotypes. Further studies revealed that ETV4/LOXL2 complex could bind NID1 promoter to mediate its demethylation, induce NID1 expression and subsequent ERK signaling pathway activation, which is required for ETV4/LOXL2-mediated EMT and metastasis of CRC. Meanwhile, the expression of ETV4 and LOXL2 were significantly negatively correlated with the methylation of NID1 promoter in clinical samples. Besides, the combined ETV4, LOXL2 and NID1 as prognostic markers is more reliable than any one alone. Taken together, in this study, we demonstrated that ETV4 played a critical role in CRC metastasis, and unraveled the novel regulatory axis of ETV4/LOXL2/NID1, which contributed to the malignant progression of CRC.
{"title":"ETV4 interacts with LOXL2 to induce epigenetic activation of NID1 during colorectal cancer progression.","authors":"Tinghui Jiang, Xin Liu, Hao Liu, Kailong Du, Yitao Wang, Hui Fan, Ying Zhang, Lin Cui, Hewei Zhang, Chundong Zhang, Yong Zhu, Zhongyu Liu, Youquan Bu, Yunlong Lei","doi":"10.7150/ijbs.116383","DOIUrl":"10.7150/ijbs.116383","url":null,"abstract":"<p><p>Colorectal cancer (CRC) is one of the most common malignant cancers worldwide and its poor prognosis is mainly caused by metastasis. Although extensive studies, the potential molecular mechanisms of CRC metastasis are not fully understood. In the present study, we found that ETV4 was remarkably upregulated in CRC and its overexpression correlated with lymph node metastasis. ETV4 could significantly promote the growth, epithelial-mesenchymal transition (EMT) and metastasis of CRC cells <i>in vitro</i> and <i>in vivo</i>. Mechanistic investigations found that LOXL2 was a novel transcriptional target and a direct interacting partner of ETV4 and was vital to ETV4-induced CRC malignant phenotypes. Further studies revealed that ETV4/LOXL2 complex could bind NID1 promoter to mediate its demethylation, induce NID1 expression and subsequent ERK signaling pathway activation, which is required for ETV4/LOXL2-mediated EMT and metastasis of CRC. Meanwhile, the expression of ETV4 and LOXL2 were significantly negatively correlated with the methylation of NID1 promoter in clinical samples. Besides, the combined ETV4, LOXL2 and NID1 as prognostic markers is more reliable than any one alone. Taken together, in this study, we demonstrated that ETV4 played a critical role in CRC metastasis, and unraveled the novel regulatory axis of ETV4/LOXL2/NID1, which contributed to the malignant progression of CRC.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":"21 15","pages":"6674-6696"},"PeriodicalIF":10.0,"publicationDate":"2025-10-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12631116/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145587223","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}
Homocysteine (Hcy) is an age-related risk factor for erectile dysfunction (ED), with enhanced vascular toxicity in middle-aged and elderly individuals. However, folate-based Hcy-lowering therapies have shown limited efficacy, necessitating a reevaluation of its age-dependent pathogenic mechanism. Here, we demonstrate that senescent endothelial cells exhibit heightened responsiveness of methionyl-tRNA synthetase 1 (MARS1) to Hcy, promoting the production of homocysteine thiolactone (HTL) and widespread N-homocysteinylation (K-Hcy) of proteins. K-Hcy, rather than acetylation, drives cytoplasmic translocation and extracellular release of high mobility group box proteins 1 and 2 (HMGB1/2), amplifying the senescence-associated secretory phenotype (SASP). Competitive inhibition of MARS1 with N-acetylcysteine (NAC) attenuates endothelial senescence and improves erectile function in middle-aged individuals with hyperhomocysteinemia by reducing HTL, rather than Hcy itself, while synergizing with tadalafil. Collectively, our findings highlight the pivotal role of the age-dependent MARS1-HTL axis in the pathogenesis of homocysteine-induced ED, offering a promising therapeutic strategy for ED in the aging population.
{"title":"N-Homocysteinylation of HMGB1/2 Promotes Corpus Cavernosum Endothelial Senescence in Erectile Dysfunction.","authors":"Peng Hu, Sen Fu, Beining Li, Xiaoyu Zhu, Bocheng Tu, Chenglin Han, Jiaxin Wang, Wenchao Xu, Xinqi Liu, Shiqing Zhu, Chengwei Wang, Zhiyao Deng, Yuxuan Deng, Sheng Xin, Jingyu Song, Jihong Liu, Kai Cui","doi":"10.7150/ijbs.119514","DOIUrl":"10.7150/ijbs.119514","url":null,"abstract":"<p><p>Homocysteine (Hcy) is an age-related risk factor for erectile dysfunction (ED), with enhanced vascular toxicity in middle-aged and elderly individuals. However, folate-based Hcy-lowering therapies have shown limited efficacy, necessitating a reevaluation of its age-dependent pathogenic mechanism. Here, we demonstrate that senescent endothelial cells exhibit heightened responsiveness of methionyl-tRNA synthetase 1 (MARS1) to Hcy, promoting the production of homocysteine thiolactone (HTL) and widespread N-homocysteinylation (K-Hcy) of proteins. K-Hcy, rather than acetylation, drives cytoplasmic translocation and extracellular release of high mobility group box proteins 1 and 2 (HMGB1/2), amplifying the senescence-associated secretory phenotype (SASP). Competitive inhibition of MARS1 with N-acetylcysteine (NAC) attenuates endothelial senescence and improves erectile function in middle-aged individuals with hyperhomocysteinemia by reducing HTL, rather than Hcy itself, while synergizing with tadalafil. Collectively, our findings highlight the pivotal role of the age-dependent MARS1-HTL axis in the pathogenesis of homocysteine-induced ED, offering a promising therapeutic strategy for ED in the aging population.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":"21 15","pages":"6723-6744"},"PeriodicalIF":10.0,"publicationDate":"2025-10-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12631104/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145587208","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}
Muscle-invasive (MI) urothelial carcinoma (UC) is a clinically challenging malignancy with a poor prognosis. Understanding the cellular dynamics that drive UC progression is critical for the development of optimized therapeutic strategies. Through integrative analysis of large-scale single-cell transcriptomic datasets from non-muscle-invasive (NMI) and MI tumours and validation with spatial transcriptomic datasets, we systematically characterized immune cell dynamics and cancer cell plasticity during UC progression. Our analysis revealed an immunosuppressive tumour microenvironment and a subset of cancer cells with upregulated major histocompatibility complex II (MHC-II) expression in MI tumours. Notably, MHC-II⁺ cancer cells were induced by interferon-γ signalling, as confirmed through in vitro experiments, and exhibited phenotypic alterations characterized by enhanced proliferative and migratory capacities. Furthermore, MHC-II⁺ cancer cells spatially colocalized with CD8⁺ T cells, regulatory T cells, and SPP1⁺ macrophages, where they engaged with inhibitory receptors on these immune cells, promoted CD8⁺ T cell exhaustion and facilitated immune evasion.
{"title":"Single-cell and spatial transcriptomic analysis reveal cellular heterogeneity and cancer cell-intrinsic major histocompatibility complex II expression in urothelial carcinoma.","authors":"Shengwei Xiong, Jian Fan, Cong Huang, Shiming He, Yuan Liang, Qi Zhang, Bin Guo, Yucai Wu, Mancheng Xia, Fangzhou Zhao, Weimin Ci, Liqun Zhou, Yanqing Gong, Xuesong Li","doi":"10.7150/ijbs.114618","DOIUrl":"10.7150/ijbs.114618","url":null,"abstract":"<p><p>Muscle-invasive (MI) urothelial carcinoma (UC) is a clinically challenging malignancy with a poor prognosis. Understanding the cellular dynamics that drive UC progression is critical for the development of optimized therapeutic strategies. Through integrative analysis of large-scale single-cell transcriptomic datasets from non-muscle-invasive (NMI) and MI tumours and validation with spatial transcriptomic datasets, we systematically characterized immune cell dynamics and cancer cell plasticity during UC progression. Our analysis revealed an immunosuppressive tumour microenvironment and a subset of cancer cells with upregulated major histocompatibility complex II (MHC-II) expression in MI tumours. Notably, MHC-II⁺ cancer cells were induced by interferon-γ signalling, as confirmed through <i>in vitro</i> experiments, and exhibited phenotypic alterations characterized by enhanced proliferative and migratory capacities. Furthermore, MHC-II⁺ cancer cells spatially colocalized with CD8⁺ T cells, regulatory T cells, and SPP1⁺ macrophages, where they engaged with inhibitory receptors on these immune cells, promoted CD8⁺ T cell exhaustion and facilitated immune evasion.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":"21 15","pages":"6649-6673"},"PeriodicalIF":10.0,"publicationDate":"2025-10-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12631105/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145587360","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 : 2025-10-20eCollection Date: 2025-01-01DOI: 10.7150/ijbs.115293
Zibin Zhan, Xuewen Liu, Min Zeng, Zehua Li, Yue Zhang, Xueyan Qiao, Xinming Li, Xianfeng Xia, Kunhao Bai, Fanhong Zeng, Yi Gao, Jun Weng
The lack of expandable human hepatocytes in vitro hampers the clinical application of the bioartificial liver. Previous studies have shown that chemical cocktails containing growth factors can support long-term expansion of hepatocytes through dedifferentiation. Here, it is revealed that chemokine (C-C motif) ligand 2 (CCL2) is a key factor in liver regeneration. CCL2 could promote the long-term expansion (over 30 passages) of human primary hepatocytes and enhancing their proliferative efficiency. Subsequently, CCL2-mediated proliferation of hepatocytes can effectively expand in vitro, and repopulate the liver of Fah-/- mice following 2-(2-nitro-4-trifluoromethylbenzyol)-1,3- cyclohexanedione (NTBC) withdrawal. Further studies revealed that CCL2-mediated hepatocyte proliferation could yield a sufficient number of highly active and well-functioning hepatocytes, crucial for supporting Bioartificial liver (BAL) therapy in treating acute liver failure (ALF) in a porcine model. Mechanically, BAL therapy effectively suppresses inflammatory responses, promotes liver regeneration, and subsequently protects extrahepatic organs, leading to improved survival rates in ALF porcine models.
{"title":"CCL2 supports human hepatocytes long-term expansion for bioartificial liver therapy to relieve acute liver failure and extrahepatic complications.","authors":"Zibin Zhan, Xuewen Liu, Min Zeng, Zehua Li, Yue Zhang, Xueyan Qiao, Xinming Li, Xianfeng Xia, Kunhao Bai, Fanhong Zeng, Yi Gao, Jun Weng","doi":"10.7150/ijbs.115293","DOIUrl":"10.7150/ijbs.115293","url":null,"abstract":"<p><p>The lack of expandable human hepatocytes in vitro hampers the clinical application of the bioartificial liver. Previous studies have shown that chemical cocktails containing growth factors can support long-term expansion of hepatocytes through dedifferentiation. Here, it is revealed that chemokine (C-C motif) ligand 2 (CCL2) is a key factor in liver regeneration. CCL2 could promote the long-term expansion (over 30 passages) of human primary hepatocytes and enhancing their proliferative efficiency. Subsequently, CCL2-mediated proliferation of hepatocytes can effectively expand in vitro, and repopulate the liver of Fah<sup>-/-</sup> mice following 2-(2-nitro-4-trifluoromethylbenzyol)-1,3- cyclohexanedione (NTBC) withdrawal. Further studies revealed that CCL2-mediated hepatocyte proliferation could yield a sufficient number of highly active and well-functioning hepatocytes, crucial for supporting Bioartificial liver (BAL) therapy in treating acute liver failure (ALF) in a porcine model. Mechanically, BAL therapy effectively suppresses inflammatory responses, promotes liver regeneration, and subsequently protects extrahepatic organs, leading to improved survival rates in ALF porcine models.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":"21 15","pages":"6759-6774"},"PeriodicalIF":10.0,"publicationDate":"2025-10-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12631109/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145586994","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}
α-hederin is a natural compound that is used to treat colorectal cancer (CRC). However, the precise anti-CRC mechanism needs to be explored further, and its direct targets have not yet been reported. In the present study, for the first time, we revealed that α-hederin directly targeted ubiquitin specific peptidase 5 (USP5), decreased its expression, weakened its interaction with signal transducer and activator of transcription 3 (STAT3), and disrupted STAT3 deubiquitination, thereby inhibiting colorectal tumorigenesis. This is particularly significant because STAT3 is a key mediator of inflammation and tumorigenesis, and targeting STAT3 deubiquitination represents a promising pathway for combating CRC; however, its deubiquitination mechanism in CRC remains unclear. USP5, a deubiquitinating enzyme (DUB) involved in inflammatory responses that is highly expressed in primary CRC tissues and promotes tumorigenesis by stabilizing tumor proteins, was identified in our study as a novel DUB of STAT3. In addition, we showed that USP5 serves as an oncogene in CRC by deubiquitinating STAT3, which contributes to CRC progression. Overall, our study provided evidence that α-hederin exhibits significant potential in suppressing colorectal tumorigenesis by disrupting USP5-mediated STAT3 deubiquitination.
α-hederin是一种用于治疗结直肠癌(CRC)的天然化合物。然而,确切的抗crc机制需要进一步探索,其直接靶点尚未报道。本研究首次发现α-hederin直接靶向泛素特异性肽酶5 (ubiquitin specific peptidase 5, USP5),降低其表达,减弱其与信号换能器和转录激活因子3 (STAT3)的相互作用,破坏STAT3的去泛素化,从而抑制结直肠肿瘤的发生。这一点尤其重要,因为STAT3是炎症和肿瘤发生的关键介质,靶向STAT3去泛素化是治疗CRC的一个有希望的途径;然而,其在结直肠癌中的去泛素化机制尚不清楚。USP5是一种参与炎症反应的去泛素化酶(DUB),在原发性结直肠癌组织中高表达,并通过稳定肿瘤蛋白促进肿瘤发生,在我们的研究中被确定为STAT3的新型DUB。此外,我们发现USP5通过去泛素化STAT3在CRC中发挥致癌基因的作用,这有助于CRC的进展。总之,我们的研究提供了证据,表明α-hederin通过破坏usp5介导的STAT3去泛素化,在抑制结直肠肿瘤发生方面具有显著的潜力。
{"title":"α-hederin Targets USP5 to Inhibit Colorectal Tumorigenesis by Disrupting STAT3 Deubiquitination.","authors":"Hui Feng, Qijuan Wang, Liu Li, Lihuiping Tao, Shuhong Zeng, Ziwen Li, Minmin Fan, Chengtao Yu, Dongdong Sun, Weixing Shen, Haibo Cheng","doi":"10.7150/ijbs.119868","DOIUrl":"10.7150/ijbs.119868","url":null,"abstract":"<p><p>α-hederin is a natural compound that is used to treat colorectal cancer (CRC). However, the precise anti-CRC mechanism needs to be explored further, and its direct targets have not yet been reported. In the present study, for the first time, we revealed that α-hederin directly targeted ubiquitin specific peptidase 5 (USP5), decreased its expression, weakened its interaction with signal transducer and activator of transcription 3 (STAT3), and disrupted STAT3 deubiquitination, thereby inhibiting colorectal tumorigenesis. This is particularly significant because STAT3 is a key mediator of inflammation and tumorigenesis, and targeting STAT3 deubiquitination represents a promising pathway for combating CRC; however, its deubiquitination mechanism in CRC remains unclear. USP5, a deubiquitinating enzyme (DUB) involved in inflammatory responses that is highly expressed in primary CRC tissues and promotes tumorigenesis by stabilizing tumor proteins, was identified in our study as a novel DUB of STAT3. In addition, we showed that USP5 serves as an oncogene in CRC by deubiquitinating STAT3, which contributes to CRC progression. Overall, our study provided evidence that α-hederin exhibits significant potential in suppressing colorectal tumorigenesis by disrupting USP5-mediated STAT3 deubiquitination.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":"21 15","pages":"6697-6722"},"PeriodicalIF":10.0,"publicationDate":"2025-10-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12631106/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145586704","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}
Radiotherapy is a primary treatment for glioblastoma (GBM), yet its effectiveness is limited by frequent recurrence due to radioresistance. Our previous studies have illustrated that GDF15 is highly expressed in radioresistant GBM cells and correlates strongly with recurrent GBM tissue. However, its role in radioresistance remained unclear. Here, we demonstrate that GDF15 promotes radioresistance by suppressing ferroptosis and altering the immune microenvironment. Mechanistically, GDF15 alleviates radiation-induced ferroptosis by stabilizing NRF2 protein through reduced ubiquitin-mediated degradation. Additionally, after radiation, GDF15 promotes M2 type macrophage infiltration, fostering an immunosuppressive microenvironment that further supports radioresistance. These findings emphasize GDF15 as a key mediator of GBM radioresistance and a potential therapeutic target to improve radiotherapy outcomes.
{"title":"GDF15 Drives Glioblastoma Radioresistance by Inhibiting Ferroptosis and Remodeling the Immune Microenvironment.","authors":"Wenqing Feng, Yantan Liu, Qinghua Zhang, Shushu Hu, Dehuang Xie, Peixin Tan, Yuan Lei, Chen Chen, Chen Ren, Shasha Du","doi":"10.7150/ijbs.115721","DOIUrl":"10.7150/ijbs.115721","url":null,"abstract":"<p><p>Radiotherapy is a primary treatment for glioblastoma (GBM), yet its effectiveness is limited by frequent recurrence due to radioresistance. Our previous studies have illustrated that GDF15 is highly expressed in radioresistant GBM cells and correlates strongly with recurrent GBM tissue. However, its role in radioresistance remained unclear. Here, we demonstrate that GDF15 promotes radioresistance by suppressing ferroptosis and altering the immune microenvironment. Mechanistically, GDF15 alleviates radiation-induced ferroptosis by stabilizing NRF2 protein through reduced ubiquitin-mediated degradation. Additionally, after radiation, GDF15 promotes M2 type macrophage infiltration, fostering an immunosuppressive microenvironment that further supports radioresistance. These findings emphasize GDF15 as a key mediator of GBM radioresistance and a potential therapeutic target to improve radiotherapy outcomes.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":"21 15","pages":"6794-6807"},"PeriodicalIF":10.0,"publicationDate":"2025-10-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12631175/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145587253","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: Gastric cancer (GC), a prevalent and life-threatening malignancy, poses significant challenges in diagnosis and prognosis due to its complex molecular pathogenesis. Identifying novel biomarkers and therapeutic targets is crucial for advancing treatment strategies and improving patient outcomes. This study investigates the role of synaptotagmin-4 (SYT4), recently identified as an oncogene, in GC development. Methods: We integrated proteomic and clinical analyses to evaluate SYT4 expression levels and their correlations with clinical features. Bioinformatic and clinicopathological assessments further validated SYT4's clinical relevance. Through comprehensive in vitro and in vivo experiments-including immunoprecipitation-mass spectrometry (IP-MS), co-immunoprecipitation (Co-IP), GST pull-down assays, and TOP/FOP luciferase reporter assays-we delineated SYT4's biological functions and interaction mechanisms. Additionally, we investigated the therapeutic potential of borussertib, a specific SYT4 inhibitor, in suppressing GC tumorigenicity. Results: SYT4 expression was significantly upregulated in GC tissues and strongly correlated with poor prognosis. Functionally, SYT4 drove cell proliferation, promoted cell cycle progression, and suppressed apoptosis in both cellular and animal models. Mechanistic investigations revealed that SYT4 directly interacts with PSMC6 via its C2B domain (amino acids 288-423), and stabilizes PSMC6 protein, thereby activating the Wnt/β-catenin signaling pathway. Notably, borussertib, a targeted SYT4 inhibitor, markedly suppressed SYT4 activity, leading to attenuated GC progression. Conclusion: Our findings demonstrate that SYT4 is a critical driver of GC progression via activation of the Wnt/β-catenin pathway. Moreover, we uncovered a novel mechanism by which borussertib selectively inhibits SYT4's oncogenic activity, providing compelling evidence for its therapeutic potential in gastric cancer treatment.
{"title":"SYT4 Interacts with PSMC6 to Facilitate Malignant Progression in Gastric Carcinoma via Activating Wnt/β-catenin Signaling.","authors":"Wen Huang, Rongkui Luo, Huimei Wang, Shuo Yang, Zixiang Yu, Yufeng Liu, Huaiyu Liang, Yanyan Shen, Xiaolei Zhang, Licheng Shen, Sujie Akesu, Chen Xu, Yingyong Hou","doi":"10.7150/ijbs.118672","DOIUrl":"10.7150/ijbs.118672","url":null,"abstract":"<p><p><b>Background:</b> Gastric cancer (GC), a prevalent and life-threatening malignancy, poses significant challenges in diagnosis and prognosis due to its complex molecular pathogenesis. Identifying novel biomarkers and therapeutic targets is crucial for advancing treatment strategies and improving patient outcomes. This study investigates the role of synaptotagmin-4 (SYT4), recently identified as an oncogene, in GC development. <b>Methods:</b> We integrated proteomic and clinical analyses to evaluate SYT4 expression levels and their correlations with clinical features. Bioinformatic and clinicopathological assessments further validated SYT4's clinical relevance. Through comprehensive <i>in vitro</i> and <i>in vivo</i> experiments-including immunoprecipitation-mass spectrometry (IP-MS), co-immunoprecipitation (Co-IP), GST pull-down assays, and TOP/FOP luciferase reporter assays-we delineated SYT4's biological functions and interaction mechanisms. Additionally, we investigated the therapeutic potential of borussertib, a specific SYT4 inhibitor, in suppressing GC tumorigenicity. <b>Results:</b> SYT4 expression was significantly upregulated in GC tissues and strongly correlated with poor prognosis. Functionally, SYT4 drove cell proliferation, promoted cell cycle progression, and suppressed apoptosis in both cellular and animal models. Mechanistic investigations revealed that SYT4 directly interacts with PSMC6 via its C2B domain (amino acids 288-423), and stabilizes PSMC6 protein, thereby activating the Wnt/β-catenin signaling pathway. Notably, borussertib, a targeted SYT4 inhibitor, markedly suppressed SYT4 activity, leading to attenuated GC progression. <b>Conclusion:</b> Our findings demonstrate that SYT4 is a critical driver of GC progression via activation of the Wnt/β-catenin pathway. Moreover, we uncovered a novel mechanism by which borussertib selectively inhibits SYT4's oncogenic activity, providing compelling evidence for its therapeutic potential in gastric cancer treatment.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":"21 15","pages":"6775-6793"},"PeriodicalIF":10.0,"publicationDate":"2025-10-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12631102/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145587290","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}