首页 > 最新文献

International Journal of Biological Sciences最新文献

英文 中文
SIRT1 in Liver Diseases: Mechanistic Insights and Therapeutic Prospects. SIRT1在肝脏疾病中的作用机制及治疗前景
IF 1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-11-05 eCollection Date: 2025-01-01 DOI: 10.7150/ijbs.117669
Fucheng Zuo, Junfa Yang, Qixiang Wu, Yaru Yang, Huan Zhou, Yuansong Sun, Tao Xu

Liver diseases present a formidable global health challenge and rank among the leading causes of morbidity and premature mortality worldwide. Silent information regulator 1 (SIRT1), a nicotinamide adenine dinucleotide (NAD⁺)-dependent histone deacetylase, has emerged as a crucial regulator of various pathophysiological processes, including metabolic homeostasis, inflammatory responses and apoptosis. This evolutionarily conserved enzyme exhibits a disease‑specific expression profile and is subject to tightly regulated mechanisms in diverse liver diseases. In recent years, accumulating evidence has highlighted the critical involvement of SIRT1 dysregulation in the pathogenesis of various liver diseases. In this review, we provide a comprehensive overview of the roles of SIRT1 in multiple liver diseases, including metabolic dysfunction-associated steatotic liver disease (MASLD), alcohol-associated liver disease (ALD), liver fibrosis, and hepatocellular carcinoma (HCC). We further explore the underlying regulatory mechanisms, aiming to establish a rigorous framework to facilitate the clinical translation of SIRT1-targeted therapeutic strategies.

肝脏疾病是一个巨大的全球健康挑战,是全世界发病率和过早死亡的主要原因之一。沉默信息调节因子1 (SIRT1)是一种烟酰胺腺嘌呤二核苷酸(NAD⁺)依赖的组蛋白去乙酰化酶,已经成为各种病理生理过程的关键调节因子,包括代谢稳态、炎症反应和细胞凋亡。这种进化上保守的酶表现出疾病特异性表达谱,并在多种肝脏疾病中受到严格调节的机制。近年来,越来越多的证据强调SIRT1失调在各种肝脏疾病的发病机制中的关键作用。在这篇综述中,我们全面概述了SIRT1在多种肝脏疾病中的作用,包括代谢功能障碍相关的脂肪变性肝病(MASLD)、酒精相关肝病(ALD)、肝纤维化和肝细胞癌(HCC)。我们进一步探索潜在的调控机制,旨在建立一个严格的框架,以促进sirt1靶向治疗策略的临床翻译。
{"title":"SIRT1 in Liver Diseases: Mechanistic Insights and Therapeutic Prospects.","authors":"Fucheng Zuo, Junfa Yang, Qixiang Wu, Yaru Yang, Huan Zhou, Yuansong Sun, Tao Xu","doi":"10.7150/ijbs.117669","DOIUrl":"10.7150/ijbs.117669","url":null,"abstract":"<p><p>Liver diseases present a formidable global health challenge and rank among the leading causes of morbidity and premature mortality worldwide. Silent information regulator 1 (SIRT1), a nicotinamide adenine dinucleotide (NAD⁺)-dependent histone deacetylase, has emerged as a crucial regulator of various pathophysiological processes, including metabolic homeostasis, inflammatory responses and apoptosis. This evolutionarily conserved enzyme exhibits a disease‑specific expression profile and is subject to tightly regulated mechanisms in diverse liver diseases. In recent years, accumulating evidence has highlighted the critical involvement of SIRT1 dysregulation in the pathogenesis of various liver diseases. In this review, we provide a comprehensive overview of the roles of SIRT1 in multiple liver diseases, including metabolic dysfunction-associated steatotic liver disease (MASLD), alcohol-associated liver disease (ALD), liver fibrosis, and hepatocellular carcinoma (HCC). We further explore the underlying regulatory mechanisms, aiming to establish a rigorous framework to facilitate the clinical translation of SIRT1-targeted therapeutic strategies.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":"21 15","pages":"7086-7103"},"PeriodicalIF":10.0,"publicationDate":"2025-11-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12631179/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145587315","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}
引用次数: 0
Proteasome: Role in T Cell Function Regulation. 蛋白酶体:在T细胞功能调节中的作用。
IF 1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-11-05 eCollection Date: 2025-01-01 DOI: 10.7150/ijbs.125134
Dongyang Tang, Xiaoran Wu, Josh Haipeng Lei, Yunfeng Qiao, Chu-Xia Deng

The proteasome plays a pivotal role in proteostasis and is deeply involved in various cellular processes. Currently, three proteasome inhibitors have been used for clinical therapies of liquid cancers with favorable efficacy, however they fail to achieve ideal efficiency in clinical trials for solid cancers without a clear clue. Recent studies have unveiled that beyond its canonical role in ubiquitin-mediated protein degradation, the proteasome also elicits a multifaceted influence on T cell fate, steering it through antigen processing, metabolic reprogramming, and the prevention of exhaustion. The proteasome inhibitors may affect tumor progression through their critical role in modulating T cell-mediated antitumor immunity, an understanding of which may solve the mystery underlying the poor efficacy of the proteasome inhibitors for solid cancers and unlock novel strategies for precision immunotherapy. This review will summarize the current knowledge of how proteasome activity weaves its threads through thymic selection, T cell aging, activation, differentiation, and immune evasion. Moreover, we will explore how cutting-edge technologies-CRISPR editing, single-cell proteomics, and AI-driven drug design can expand the application of the proteasome inhibitors in the treatment of cancer and autoimmune diseases.

蛋白酶体在蛋白质平衡中起着关键作用,并深入参与各种细胞过程。目前,已有三种蛋白酶体抑制剂用于液体癌的临床治疗,疗效良好,但在实体癌的临床试验中未能达到理想效果,缺乏明确的线索。最近的研究表明,除了在泛素介导的蛋白质降解中发挥典型作用外,蛋白酶体还对T细胞命运产生多方面的影响,通过抗原加工、代谢重编程和防止衰竭来引导T细胞。蛋白酶体抑制剂可能通过其在调节T细胞介导的抗肿瘤免疫中的关键作用来影响肿瘤进展,对其的理解可能解决蛋白酶体抑制剂对实体癌疗效差的奥秘,并为精确免疫治疗提供新的策略。本文将对蛋白酶体活性如何在胸腺选择、T细胞老化、活化、分化和免疫逃避中编织其线索进行综述。此外,我们将探索尖端技术——crispr编辑、单细胞蛋白质组学和人工智能驱动的药物设计——如何扩大蛋白酶体抑制剂在癌症和自身免疫性疾病治疗中的应用。
{"title":"Proteasome: Role in T Cell Function Regulation.","authors":"Dongyang Tang, Xiaoran Wu, Josh Haipeng Lei, Yunfeng Qiao, Chu-Xia Deng","doi":"10.7150/ijbs.125134","DOIUrl":"10.7150/ijbs.125134","url":null,"abstract":"<p><p>The proteasome plays a pivotal role in proteostasis and is deeply involved in various cellular processes. Currently, three proteasome inhibitors have been used for clinical therapies of liquid cancers with favorable efficacy, however they fail to achieve ideal efficiency in clinical trials for solid cancers without a clear clue. Recent studies have unveiled that beyond its canonical role in ubiquitin-mediated protein degradation, the proteasome also elicits a multifaceted influence on T cell fate, steering it through antigen processing, metabolic reprogramming, and the prevention of exhaustion. The proteasome inhibitors may affect tumor progression through their critical role in modulating T cell-mediated antitumor immunity, an understanding of which may solve the mystery underlying the poor efficacy of the proteasome inhibitors for solid cancers and unlock novel strategies for precision immunotherapy. This review will summarize the current knowledge of how proteasome activity weaves its threads through thymic selection, T cell aging, activation, differentiation, and immune evasion. Moreover, we will explore how cutting-edge technologies-CRISPR editing, single-cell proteomics, and AI-driven drug design can expand the application of the proteasome inhibitors in the treatment of cancer and autoimmune diseases.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":"21 15","pages":"7104-7117"},"PeriodicalIF":10.0,"publicationDate":"2025-11-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12631184/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145587201","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}
引用次数: 0
Activation of GSDME by all-trans-retinal increases sensitivity to photoreceptor ferroptosis. 全反式视网膜激活GSDME增加对光感受器铁下垂的敏感性。
IF 1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-10-27 eCollection Date: 2025-01-01 DOI: 10.7150/ijbs.114187
Bo Yang, Kunhuan Yang, Yuling Chen, Ruitong Xi, Jiahuai Han, Shiying Li, Jingmeng Chen, Yalin Wu

Impaired clearance of all-trans-retinal (atRAL) due to visual cycle dysfunction contributes to photoreceptor atrophy, a key pathological hallmark of Stargardt disease type 1 (STGD1) and dry age-related macular degeneration (AMD). Prior studies have shown that light-induced atRAL accumulation promotes ferroptosis and activates gasdermin E (GSDME) in retinal photoreceptors of Abca4-/-Rdh8-/- mice, a model for STGD1 and dry AMD that exhibits visual cycle disorders. However, the role of GSDME in photoreceptor ferroptosis remains unclear. In this study, we revealed that GSDME activation by atRAL triggered photoreceptor ferroptosis and retinal atrophy via mitochondrial damage and oxidative stress. Knocking out GSDME significantly attenuated light-induced photoreceptor ferroptosis and retinal degeneration in Abca4-/-Rdh8-/- mice. Moreover, deleting the Gsdme gene in photoreceptor cells prevented atRAL-induced ferroptosis by inhibiting mitochondrial reactive oxygen species (mitoROS) production, iron overload, and lipid peroxidation. Notably, treatment with the mitoROS scavenger MitoTEMPO mitigated ferroptosis in atRAL-loaded photoreceptor cells and dramatically relieved photoreceptor ferroptosis and retinal degeneration in light-exposed Abca4-/-Rdh8-/- mice. We found that both GSDME elimination and MitoTEMPO treatment repressed atRAL-induced photoreceptor ferroptosis and retinal atrophy by inactivating the mitoROS-induced oxidative stress. In conclusion, GSDME-mediated photoreceptor ferroptosis is crucial for inducing structural and functional damage of the retina in retinopathies caused by atRAL accumulation, thereby providing new therapeutic insights for the prevention and treatment of STGD1 and dry AMD.

由于视觉周期功能障碍导致的全反式视网膜(atRAL)清除受损导致光感受器萎缩,这是Stargardt病1型(STGD1)和干性年龄相关性黄斑变性(AMD)的一个关键病理标志。先前的研究表明,光诱导的atRAL积累促进了Abca4-/- rdh8 -/-小鼠视网膜光感受器中的铁凋亡并激活了气皮蛋白E (GSDME), Abca4-/- rdh8 -/-小鼠是STGD1和干性AMD表现出视觉周期障碍的模型。然而,GSDME在光感受器铁下垂中的作用尚不清楚。在这项研究中,我们发现atRAL激活GSDME通过线粒体损伤和氧化应激引发光感受器铁下垂和视网膜萎缩。敲除GSDME可显著减弱Abca4-/- rdh8 -/-小鼠光致光感受器铁下垂和视网膜变性。此外,删除光感受器细胞中的Gsdme基因可以通过抑制线粒体活性氧(mitoROS)的产生、铁过载和脂质过氧化来防止atral诱导的铁死亡。值得注意的是,使用mitoROS清扫剂MitoTEMPO治疗可减轻atral负载的光感受器细胞的铁下垂,并显着减轻光暴露的Abca4-/- rdh8 -/-小鼠的光感受器铁下垂和视网膜变性。我们发现GSDME消除和MitoTEMPO治疗均通过灭活mitoros诱导的氧化应激来抑制atral诱导的光感受器铁下垂和视网膜萎缩。综上所述,gsdme介导的光感受器ferroptosis在atRAL积累引起的视网膜病变中诱导视网膜结构和功能损伤至关重要,从而为STGD1和干性AMD的预防和治疗提供了新的治疗见解。
{"title":"Activation of GSDME by all-<i>trans</i>-retinal increases sensitivity to photoreceptor ferroptosis.","authors":"Bo Yang, Kunhuan Yang, Yuling Chen, Ruitong Xi, Jiahuai Han, Shiying Li, Jingmeng Chen, Yalin Wu","doi":"10.7150/ijbs.114187","DOIUrl":"10.7150/ijbs.114187","url":null,"abstract":"<p><p>Impaired clearance of all-<i>trans</i>-retinal (atRAL) due to visual cycle dysfunction contributes to photoreceptor atrophy, a key pathological hallmark of Stargardt disease type 1 (STGD1) and dry age-related macular degeneration (AMD). Prior studies have shown that light-induced atRAL accumulation promotes ferroptosis and activates gasdermin E (GSDME) in retinal photoreceptors of <i>Abca4<sup>-/-</sup>Rdh8<sup>-/-</sup></i> mice, a model for STGD1 and dry AMD that exhibits visual cycle disorders. However, the role of GSDME in photoreceptor ferroptosis remains unclear. In this study, we revealed that GSDME activation by atRAL triggered photoreceptor ferroptosis and retinal atrophy via mitochondrial damage and oxidative stress. Knocking out GSDME significantly attenuated light-induced photoreceptor ferroptosis and retinal degeneration in <i>Abca4<sup>-/-</sup>Rdh8<sup>-/-</sup></i> mice. Moreover, deleting the <i>Gsdme</i> gene in photoreceptor cells prevented atRAL-induced ferroptosis by inhibiting mitochondrial reactive oxygen species (mitoROS) production, iron overload, and lipid peroxidation. Notably, treatment with the mitoROS scavenger MitoTEMPO mitigated ferroptosis in atRAL-loaded photoreceptor cells and dramatically relieved photoreceptor ferroptosis and retinal degeneration in light-exposed <i>Abca4<sup>-/-</sup>Rdh8<sup>-/-</sup></i> mice. We found that both GSDME elimination and MitoTEMPO treatment repressed atRAL-induced photoreceptor ferroptosis and retinal atrophy by inactivating the mitoROS-induced oxidative stress. In conclusion, GSDME-mediated photoreceptor ferroptosis is crucial for inducing structural and functional damage of the retina in retinopathies caused by atRAL accumulation, thereby providing new therapeutic insights for the prevention and treatment of STGD1 and dry AMD.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":"21 15","pages":"7029-7042"},"PeriodicalIF":10.0,"publicationDate":"2025-10-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12631243/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145587046","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}
引用次数: 0
GDF10 attenuates MASH progression by restoring quiescent hepatic stellate cells via competitive inhibition of TGF-β/SMAD2 signaling. GDF10通过竞争性抑制TGF-β/SMAD2信号通路,恢复静止的肝星状细胞,从而减缓MASH进展。
IF 1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-10-27 eCollection Date: 2025-01-01 DOI: 10.7150/ijbs.123784
Yajie Peng, Hongyan Lei, Jiahui Zhao, Huajuan Wang, Zheng Luo, Dixin Wang, Shujun Shi, Tianyi Wang, Jin Li, Zhiqing Pang, Bo Wang, Xuelian Xiong

Liver fibrosis has emerged as the primary determinant of outcomes in metabolic dysfunction-associated steatohepatitis (MASH). Quiescent hepatic stellate cells (HSCs) differentiate into activated HSCs or myofibroblasts, which drives liver fibrosis and contribute to the progressive loss of hepatic function. MASH with progressive fibrosis lacks effective therapies due to incomplete understanding of HSCs regulation. Here, we identify growth differentiation factor 10 (GDF10) as a master regulator of HSCs quiescence that ameliorates fibrosis through shifting HSC functions to restore HSC balance of transcriptional and metabolic reprogramming. Single-cell RNA sequencing revealed HSC-specific Gdf10 expression inversely correlated with fibrotic activation. In murine models of diet-induced MASH and CCl4-induced fibrosis, AAV-mediated Gdf10 overexpression reduced collagen deposition, serum ALT/AST, and fibrogenic gene expression without perturbing glucose or lipid metabolism. Mechanistically, GDF10 competitively bound TGF-β receptor 2 (TβR2), inhibiting SMAD2/3 phosphorylation and nuclear translocation, ultimately suppressing TGFβ1-driven extracellular matrix production, and reversing the activated HSCs phenotype and their hypermetabolic states. Leveraging this pathway, we developed liver-targeted lipid nanoparticles (LNPs) encapsulating mGdf10 mRNA, which selectively delivered Gdf10 to HSCs, reversed fibrosis in multiple animal models. Clinically, GDF10 expression correlated with fibrosis severity in human cirrhotic livers. Our findings establish GDF10 as a dual-function modulator of TGF-β signaling and HSC metabolism, offering a targeted therapeutic strategy for liver fibrosis.

肝纤维化已成为代谢功能障碍相关脂肪性肝炎(MASH)预后的主要决定因素。静止的肝星状细胞(hsc)分化为活化的hsc或肌成纤维细胞,导致肝纤维化并导致肝功能的进行性丧失。由于对造血干细胞调控的不完全了解,进行性纤维化的MASH缺乏有效的治疗方法。在这里,我们确定生长分化因子10 (GDF10)是HSC静止的主要调节因子,通过改变HSC功能来改善纤维化,从而恢复HSC转录和代谢重编程的平衡。单细胞RNA测序显示hsc特异性Gdf10表达与纤维化激活呈负相关。在小鼠饮食诱导的MASH和ccl4诱导的纤维化模型中,aav介导的Gdf10过表达减少了胶原沉积、血清ALT/AST和纤维化基因表达,而不干扰葡萄糖或脂质代谢。机制上,GDF10竞争性结合TGF-β受体2 (t -β r2),抑制SMAD2/3磷酸化和核易位,最终抑制TGF-β 1驱动的细胞外基质生成,逆转活化的hsc表型及其高代谢状态。利用这一途径,我们开发了包裹mdf10 mRNA的肝脏靶向脂质纳米颗粒(LNPs),可选择性地将Gdf10递送至hsc,在多种动物模型中逆转纤维化。在临床上,GDF10的表达与肝硬化患者的纤维化严重程度相关。我们的研究结果证实了GDF10作为TGF-β信号和HSC代谢的双功能调节剂,为肝纤维化提供了靶向治疗策略。
{"title":"GDF10 attenuates MASH progression by restoring quiescent hepatic stellate cells via competitive inhibition of TGF-β/SMAD2 signaling.","authors":"Yajie Peng, Hongyan Lei, Jiahui Zhao, Huajuan Wang, Zheng Luo, Dixin Wang, Shujun Shi, Tianyi Wang, Jin Li, Zhiqing Pang, Bo Wang, Xuelian Xiong","doi":"10.7150/ijbs.123784","DOIUrl":"10.7150/ijbs.123784","url":null,"abstract":"<p><p>Liver fibrosis has emerged as the primary determinant of outcomes in metabolic dysfunction-associated steatohepatitis (MASH). Quiescent hepatic stellate cells (HSCs) differentiate into activated HSCs or myofibroblasts, which drives liver fibrosis and contribute to the progressive loss of hepatic function. MASH with progressive fibrosis lacks effective therapies due to incomplete understanding of HSCs regulation. Here, we identify growth differentiation factor 10 (GDF10) as a master regulator of HSCs quiescence that ameliorates fibrosis through shifting HSC functions to restore HSC balance of transcriptional and metabolic reprogramming. Single-cell RNA sequencing revealed HSC-specific <i>Gdf10</i> expression inversely correlated with fibrotic activation. In murine models of diet-induced MASH and CCl4-induced fibrosis, AAV-mediated <i>Gdf10</i> overexpression reduced collagen deposition, serum ALT/AST, and fibrogenic gene expression without perturbing glucose or lipid metabolism. Mechanistically, GDF10 competitively bound TGF-β receptor 2 (TβR2), inhibiting SMAD2/3 phosphorylation and nuclear translocation, ultimately suppressing TGFβ1-driven extracellular matrix production, and reversing the activated HSCs phenotype and their hypermetabolic states. Leveraging this pathway, we developed liver-targeted lipid nanoparticles (LNPs) encapsulating <i>mGdf10</i> mRNA, which selectively delivered <i>Gdf10</i> to HSCs, reversed fibrosis in multiple animal models. Clinically, <i>GDF10</i> expression correlated with fibrosis severity in human cirrhotic livers. Our findings establish GDF10 as a dual-function modulator of TGF-β signaling and HSC metabolism, offering a targeted therapeutic strategy for liver fibrosis.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":"21 15","pages":"6997-7012"},"PeriodicalIF":10.0,"publicationDate":"2025-10-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12631229/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145587218","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}
引用次数: 0
Combination of FTO and BTK inhibitors synergistically suppresses the malignancy of breast cancer cells. FTO和BTK抑制剂联合使用可协同抑制乳腺癌细胞的恶性肿瘤。
IF 1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-10-27 eCollection Date: 2025-01-01 DOI: 10.7150/ijbs.117999
Abdulaziz Ahmed A Saad, Lichen Ge, Haoran Wang, Yan Xia, Jianing Li, Shiyao Qiao, Cheng Yi, Xiansong Wang, Zhaotong Wang, Dan Zhou, Hongsheng Wang

Despite significant progress in breast cancer treatment, more effective methods for its clinical management are still needed. Our data identified that fat mass and obesity-associated protein (FTO), an N 6-methyladenosine (m6A) demethylase, is highly expressed in breast cancer and promotes tumorigenesis. Inhibiting FTO can suppress the proliferation and metastasis of breast cancer, while its efficacy needs to be further improved. Through screening with 27 clinically approved targeted therapy drugs, we discovered that ibrutinib, a BTK inhibitor, shows the highest cell death rate and lowest combination index (CI). This combination demonstrates a potent synergistic effect in the malignancy of breast cancer and its lung metastasis. RNA-seq showed that the oncogenic pathways regulated by c-Myc and E2F1 were among the most down-regulated in cells treated with FTO inhibitor and ibrutinib. Furthermore, this combination decreases the expression of both c-Myc and E2F1. Contrarily, overexpressing c-Myc and E2F1 counteracts this antitumor effectiveness. Mechanistically, this combination inhibits c-Myc and E2F1 expression by increasing m6A modification of their mRNAs and reducing their mRNA stability. In mouse models of cancer, combining FTO knockdown with ibrutinib markedly suppressed tumor growth, decreased metastasis, and improved survival. Collectively, the combined inhibition of FTO and BTK exhibited substantial synergistic anticancer effects in breast cancer. Our findings advocate for the evaluation of this combination in clinical trials.

尽管乳腺癌治疗取得了重大进展,但仍需要更有效的临床管理方法。我们的数据表明,脂肪质量和肥胖相关蛋白(FTO),一种n6 -甲基腺苷(m6A)去甲基化酶,在乳腺癌中高度表达并促进肿瘤发生。抑制FTO可以抑制乳腺癌的增殖和转移,但其疗效有待进一步提高。通过对27种临床批准的靶向治疗药物的筛选,我们发现BTK抑制剂ibrutinib的细胞死亡率最高,联合指数(CI)最低。这种组合在乳腺癌恶性肿瘤及其肺转移中显示出强有力的协同作用。RNA-seq显示,在FTO抑制剂和伊鲁替尼处理的细胞中,c-Myc和E2F1调控的致癌途径下调最多。此外,这种组合降低了c-Myc和E2F1的表达。相反,过表达c-Myc和E2F1会抵消这种抗肿瘤效果。从机制上讲,这种组合通过增加m6A修饰它们的mRNA并降低它们的mRNA稳定性来抑制c-Myc和E2F1的表达。在小鼠癌症模型中,FTO敲除与伊鲁替尼联合使用可显著抑制肿瘤生长,减少转移,提高生存率。总的来说,FTO和BTK的联合抑制在乳腺癌中表现出实质性的协同抗癌作用。我们的研究结果支持在临床试验中对这种组合进行评估。
{"title":"Combination of FTO and BTK inhibitors synergistically suppresses the malignancy of breast cancer cells.","authors":"Abdulaziz Ahmed A Saad, Lichen Ge, Haoran Wang, Yan Xia, Jianing Li, Shiyao Qiao, Cheng Yi, Xiansong Wang, Zhaotong Wang, Dan Zhou, Hongsheng Wang","doi":"10.7150/ijbs.117999","DOIUrl":"10.7150/ijbs.117999","url":null,"abstract":"<p><p>Despite significant progress in breast cancer treatment, more effective methods for its clinical management are still needed. Our data identified that fat mass and obesity-associated protein (FTO), an <i>N</i> <sup>6</sup>-methyladenosine (m<sup>6</sup>A) demethylase, is highly expressed in breast cancer and promotes tumorigenesis. Inhibiting FTO can suppress the proliferation and metastasis of breast cancer, while its efficacy needs to be further improved. Through screening with 27 clinically approved targeted therapy drugs, we discovered that ibrutinib, a BTK inhibitor, shows the highest cell death rate and lowest combination index (CI). This combination demonstrates a potent synergistic effect in the malignancy of breast cancer and its lung metastasis. RNA-seq showed that the oncogenic pathways regulated by c-Myc and E2F1 were among the most down-regulated in cells treated with FTO inhibitor and ibrutinib. Furthermore, this combination decreases the expression of both c-Myc and E2F1. Contrarily, overexpressing c-Myc and E2F1 counteracts this antitumor effectiveness. Mechanistically, this combination inhibits c-Myc and E2F1 expression by increasing m<sup>6</sup>A modification of their mRNAs and reducing their mRNA stability. In mouse models of cancer, combining FTO knockdown with ibrutinib markedly suppressed tumor growth, decreased metastasis, and improved survival. Collectively, the combined inhibition of FTO and BTK exhibited substantial synergistic anticancer effects in breast cancer. Our findings advocate for the evaluation of this combination in clinical trials.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":"21 15","pages":"7063-7085"},"PeriodicalIF":10.0,"publicationDate":"2025-10-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12631244/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145587265","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}
引用次数: 0
Synergistic Anti-Tumor Effects of Sulfatinib and Kaempferol on Pancreatic Neuroendocrine Tumors via CALCA-mediated PI3K/AKT/mTOR Pathway. 舒法替尼和山奈酚通过calca介导的PI3K/AKT/mTOR通路对胰腺神经内分泌肿瘤的协同抗肿瘤作用。
IF 1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-10-27 eCollection Date: 2025-01-01 DOI: 10.7150/ijbs.119176
Lingyi Chen, Pengfei Liu, Fengjuan Chen, Bingyan Xue, Xu Han, Lijun Yan, Jianan Bai, Xiaoya Li, Min Liu, Ye Tian, Mujie Ye, Qiyun Tang

Pancreatic neuroendocrine tumors (pNETs) represent a diverse category of neoplasms originating from pancreatic neuroendocrine cells. Although these tumors generally exhibit a relatively indolent nature, they often metastasize early in their course, significantly affecting patient outcomes. Sulfatinib (SULF) is associated with considerable toxicity and resistance challenges, leading to many patients failing to achieve long-term disease management. In contrast, Kaempferol (KMP), a naturally occurring phytochemical, has shown considerable promise in anti-tumor treatments. Our study revealed that the combination of SULF and low-dose KMP enhances the sensitivity of pNET cells to SULF. Moreover, this combination demonstrated a synergistic effect on angiogenesis inhibition, observed in both in vitro and in vivo environments. Additionally, we confirmed this synergistic anti-tumor effect using a subcutaneous tumor model of pNETs. Transcriptome sequencing identified CALCA as a key molecule in the synergistic inhibition of pNETs proliferation by SULF and KMP. In summary, our findings provide novel insights into combination therapy for pNETs while elucidating the mechanistic role of CALCA in the modulation of angiogenesis. This research establishes a foundation for the development of vascular-targeted combination therapeutic strategies for the treatment of pNETs.

胰腺神经内分泌肿瘤(pNETs)是一类起源于胰腺神经内分泌细胞的肿瘤。虽然这些肿瘤通常表现出相对惰性的性质,但它们经常在病程早期转移,显著影响患者的预后。Sulfatinib (SULF)具有相当大的毒性和耐药性挑战,导致许多患者无法实现长期疾病管理。相反,山奈酚(KMP),一种天然存在的植物化学物质,在抗肿瘤治疗中显示出相当大的希望。我们的研究表明,联合使用SULF和低剂量KMP可以增强pNET细胞对SULF的敏感性。此外,这种组合在体外和体内环境中都显示出对血管生成抑制的协同作用。此外,我们用pNETs皮下肿瘤模型证实了这种协同抗肿瘤作用。转录组测序发现CALCA是SULF和KMP协同抑制pNETs增殖的关键分子。总之,我们的研究结果为pNETs的联合治疗提供了新的见解,同时阐明了CALCA在血管生成调节中的机制作用。本研究为开发血管靶向联合治疗pNETs的策略奠定了基础。
{"title":"Synergistic Anti-Tumor Effects of Sulfatinib and Kaempferol on Pancreatic Neuroendocrine Tumors via CALCA-mediated PI3K/AKT/mTOR Pathway.","authors":"Lingyi Chen, Pengfei Liu, Fengjuan Chen, Bingyan Xue, Xu Han, Lijun Yan, Jianan Bai, Xiaoya Li, Min Liu, Ye Tian, Mujie Ye, Qiyun Tang","doi":"10.7150/ijbs.119176","DOIUrl":"10.7150/ijbs.119176","url":null,"abstract":"<p><p>Pancreatic neuroendocrine tumors (pNETs) represent a diverse category of neoplasms originating from pancreatic neuroendocrine cells. Although these tumors generally exhibit a relatively indolent nature, they often metastasize early in their course, significantly affecting patient outcomes. Sulfatinib (SULF) is associated with considerable toxicity and resistance challenges, leading to many patients failing to achieve long-term disease management. In contrast, Kaempferol (KMP), a naturally occurring phytochemical, has shown considerable promise in anti-tumor treatments. Our study revealed that the combination of SULF and low-dose KMP enhances the sensitivity of pNET cells to SULF. Moreover, this combination demonstrated a synergistic effect on angiogenesis inhibition, observed in both <i>in vitro</i> and <i>in vivo</i> environments. Additionally, we confirmed this synergistic anti-tumor effect using a subcutaneous tumor model of pNETs. Transcriptome sequencing identified CALCA as a key molecule in the synergistic inhibition of pNETs proliferation by SULF and KMP. In summary, our findings provide novel insights into combination therapy for pNETs while elucidating the mechanistic role of CALCA in the modulation of angiogenesis. This research establishes a foundation for the development of vascular-targeted combination therapeutic strategies for the treatment of pNETs.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":"21 15","pages":"7043-7062"},"PeriodicalIF":10.0,"publicationDate":"2025-10-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12631220/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145587333","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}
引用次数: 0
Venous Malformations: Unraveling Latest Mechanisms and Bridging Gaps in Targeted Therapy Development. 静脉畸形:揭示靶向治疗发展的最新机制和弥合差距。
IF 1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-10-27 eCollection Date: 2025-01-01 DOI: 10.7150/ijbs.122569
Xuan Jiang, Li Hu, Jiayi Lai, Shengfang Ge, Hui Chen, Xi Yang, Xiaoxi Lin

The vascular system plays a crucial role in maintaining homeostasis, ensuring the supply of oxygen and nutrients to tissues, while facilitating the removal of metabolic waste. Additionally, it contributes to immune defense, temperature regulation, and the transport of hormones and signaling molecules. Vascular anomaly (VA) arises due to developmental abnormalities or functional defects in the vessels. This review describes venous malformations (VM), a rare disorder predominantly caused by somatic mutations. Advances in recent research have substantially improved our understanding of the molecular mechanisms underlying these malformations, largely through the identification of their genetic origins and the study of animal models and endothelial cells derived from patients. Most of the somatic mutations associated with venous malformations affect genes within oncogenic growth factor signaling pathways, making it possible to repurpose certain cancer therapies to treat these VAs. This article summarizes the key molecular findings and explores emerging therapeutic strategies aimed at novel targets.

血管系统在维持体内平衡,确保组织的氧气和营养供应,同时促进代谢废物的清除方面起着至关重要的作用。此外,它还有助于免疫防御、温度调节、激素和信号分子的运输。血管异常(VA)是由血管发育异常或功能缺陷引起的。这篇综述描述静脉畸形(VM),一种罕见的疾病,主要是由体细胞突变引起的。最近的研究进展极大地提高了我们对这些畸形的分子机制的理解,主要是通过鉴定其遗传起源以及动物模型和来自患者的内皮细胞的研究。大多数与静脉畸形相关的体细胞突变影响致癌生长因子信号通路内的基因,这使得重新利用某些癌症疗法来治疗这些静脉畸形成为可能。本文总结了关键的分子发现,并探讨了针对新靶点的新兴治疗策略。
{"title":"Venous Malformations: Unraveling Latest Mechanisms and Bridging Gaps in Targeted Therapy Development.","authors":"Xuan Jiang, Li Hu, Jiayi Lai, Shengfang Ge, Hui Chen, Xi Yang, Xiaoxi Lin","doi":"10.7150/ijbs.122569","DOIUrl":"10.7150/ijbs.122569","url":null,"abstract":"<p><p>The vascular system plays a crucial role in maintaining homeostasis, ensuring the supply of oxygen and nutrients to tissues, while facilitating the removal of metabolic waste. Additionally, it contributes to immune defense, temperature regulation, and the transport of hormones and signaling molecules. Vascular anomaly (VA) arises due to developmental abnormalities or functional defects in the vessels. This review describes venous malformations (VM), a rare disorder predominantly caused by somatic mutations. Advances in recent research have substantially improved our understanding of the molecular mechanisms underlying these malformations, largely through the identification of their genetic origins and the study of animal models and endothelial cells derived from patients. Most of the somatic mutations associated with venous malformations affect genes within oncogenic growth factor signaling pathways, making it possible to repurpose certain cancer therapies to treat these VAs. This article summarizes the key molecular findings and explores emerging therapeutic strategies aimed at novel targets.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":"21 15","pages":"7013-7028"},"PeriodicalIF":10.0,"publicationDate":"2025-10-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12631228/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145587322","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}
引用次数: 0
Demystifying metabolic‒immune crosstalk: how amino acid metabolic reprogramming shapes the malignant phenotype and macrophage polarization of biliary and pancreatic tumors. 揭开代谢-免疫串扰的神秘:氨基酸代谢重编程如何塑造胆道和胰腺肿瘤的恶性表型和巨噬细胞极化。
IF 1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-10-27 eCollection Date: 2025-01-01 DOI: 10.7150/ijbs.122325
Jinglei Zhang, Zhuohuan Chu, Jiawen Li, Lu Xie, Cong Ding, Zihui An, Xiang Wang, Hangbin Jin, Xiaofeng Zhang, Qiang Liu, Jianfeng Yang

Biliary and pancreatic malignant tumors refer to biliary tract carcinoma (BTC) and pancreatic cancer (PC), among which BTC mainly includes cholangiocarcinoma (CCA) and gallbladder cancer (GBC), and their prognosis is poor because of the lack of effective early diagnostic methods. Although surgical resection is the preferred method for a cure, treatment options are limited for patients with advanced tumors. Therefore, the exploration of other new treatment methods is urgently needed. Currently, metabolic reprogramming is a key mechanism in the process of tumor development and progression and is closely related to cancer cell proliferation, metastasis and drug resistance. As an indispensable part of metabolic reprogramming in tumor cells, amino acid (AA) metabolic reprogramming provides an energy source for tumor cells and participates in regulating the tumor microenvironment (TME). Moreover, as important intrinsic myeloid cells, macrophages play indispensable physiological roles in malignant tumor progression. In the TME, tumor cells can not only induce peripheral immune tolerance by releasing extracellular signals but also compete with tumor-associated macrophages (TAMs) for AAs and release the resulting downstream metabolites into the TME, directly targeting and damaging immune cells and influencing macrophage polarization. Consequently, a more profound understanding of the function of AA metabolic reprogramming in biliopancreatic malignancies and their associated macrophage polarization holds the potential to facilitate the development of effective strategies for early diagnosis, prognostic assessment and targeted therapy in patients with biliopancreatic malignancies. In this paper, we review the impact of AA metabolic reprogramming on the occurrence and development of biliary and pancreatic malignant tumors, summarize the relevant mechanisms of AA metabolic reprogramming on the polarization of TAMs, and provide new therapeutic targets for AA metabolic therapies and immunotherapies for biliary and pancreatic malignant tumors.

胆道胰腺恶性肿瘤是指胆道癌(BTC)和胰腺癌(PC),其中BTC主要包括胆管癌(CCA)和胆囊癌(GBC),由于缺乏有效的早期诊断方法,其预后较差。虽然手术切除是治愈的首选方法,但晚期肿瘤患者的治疗选择有限。因此,迫切需要探索其他新的治疗方法。目前,代谢重编程是肿瘤发生发展过程中的关键机制,与癌细胞增殖、转移和耐药密切相关。氨基酸(AA)代谢重编程是肿瘤细胞代谢重编程的重要组成部分,为肿瘤细胞提供能量来源,参与肿瘤微环境(TME)的调节。巨噬细胞作为重要的内在髓系细胞,在恶性肿瘤的发展过程中发挥着不可或缺的生理作用。在TME中,肿瘤细胞不仅可以通过释放细胞外信号诱导外周免疫耐受,还可以与肿瘤相关巨噬细胞(tumor-associated macrophages, tam)竞争AAs,并将其下游代谢物释放到TME中,直接靶向和损伤免疫细胞,影响巨噬细胞极化。因此,更深入地了解AA代谢重编程在胆道胰腺恶性肿瘤及其相关巨噬细胞极化中的功能,可能有助于制定有效的策略,用于胆道胰腺恶性肿瘤患者的早期诊断、预后评估和靶向治疗。本文综述了AA代谢重编程对胆道和胰腺恶性肿瘤发生发展的影响,总结了AA代谢重编程对TAMs极化的相关机制,为胆道和胰腺恶性肿瘤的AA代谢治疗和免疫治疗提供新的治疗靶点。
{"title":"Demystifying metabolic‒immune crosstalk: how amino acid metabolic reprogramming shapes the malignant phenotype and macrophage polarization of biliary and pancreatic tumors.","authors":"Jinglei Zhang, Zhuohuan Chu, Jiawen Li, Lu Xie, Cong Ding, Zihui An, Xiang Wang, Hangbin Jin, Xiaofeng Zhang, Qiang Liu, Jianfeng Yang","doi":"10.7150/ijbs.122325","DOIUrl":"10.7150/ijbs.122325","url":null,"abstract":"<p><p>Biliary and pancreatic malignant tumors refer to biliary tract carcinoma (BTC) and pancreatic cancer (PC), among which BTC mainly includes cholangiocarcinoma (CCA) and gallbladder cancer (GBC), and their prognosis is poor because of the lack of effective early diagnostic methods. Although surgical resection is the preferred method for a cure, treatment options are limited for patients with advanced tumors. Therefore, the exploration of other new treatment methods is urgently needed. Currently, metabolic reprogramming is a key mechanism in the process of tumor development and progression and is closely related to cancer cell proliferation, metastasis and drug resistance. As an indispensable part of metabolic reprogramming in tumor cells, amino acid (AA) metabolic reprogramming provides an energy source for tumor cells and participates in regulating the tumor microenvironment (TME). Moreover, as important intrinsic myeloid cells, macrophages play indispensable physiological roles in malignant tumor progression. In the TME, tumor cells can not only induce peripheral immune tolerance by releasing extracellular signals but also compete with tumor-associated macrophages (TAMs) for AAs and release the resulting downstream metabolites into the TME, directly targeting and damaging immune cells and influencing macrophage polarization. Consequently, a more profound understanding of the function of AA metabolic reprogramming in biliopancreatic malignancies and their associated macrophage polarization holds the potential to facilitate the development of effective strategies for early diagnosis, prognostic assessment and targeted therapy in patients with biliopancreatic malignancies. In this paper, we review the impact of AA metabolic reprogramming on the occurrence and development of biliary and pancreatic malignant tumors, summarize the relevant mechanisms of AA metabolic reprogramming on the polarization of TAMs, and provide new therapeutic targets for AA metabolic therapies and immunotherapies for biliary and pancreatic malignant tumors.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":"21 15","pages":"6952-6977"},"PeriodicalIF":10.0,"publicationDate":"2025-10-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12631177/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145587195","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}
引用次数: 0
Metabolic Reprogramming in Urological Tumors: New Perspectives from Tumor Metabolic Phenotypes to Therapeutic Targets. 泌尿系统肿瘤的代谢重编程:从肿瘤代谢表型到治疗靶点的新视角。
IF 1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-10-27 eCollection Date: 2025-01-01 DOI: 10.7150/ijbs.123647
Zhuohang Li, Lin Yang, Weijia Li, Wenxue Huang, Cunzhen Ma, Boyuan Sun, Xunguo Yang, Haoxiang Xu, Zhibiao Li, Jie Zhao, Bisheng Cheng, Peng Wu

Metabolic reprogramming is a hallmark of cancer, enabling tumor cells to sustain growth, evade immune surveillance, and resist therapy. Urological tumors, including prostate, bladder, and renal cancers, exhibit distinct metabolic phenotypes driven by their unique tumor microenvironments and oncogenic pathways. This review explores the emerging landscape of tumor metabolism in urological cancers, highlighting key metabolic pathways such as glycolysis, lipid metabolism, amino acid metabolism, and redox balance. We discuss how these pathways are intricately linked to tumor progression, therapeutic resistance, and immune evasion. Furthermore, we examine novel therapeutic strategies targeting metabolic vulnerabilities, including metabolic enzyme inhibitors, synthetic lethality approaches, and metabolic modulation to enhance immunotherapy. By integrating advances in multi-omics technologies and preclinical models, we propose a framework for translating metabolic research into clinical applications. This review aims to provide a comprehensive overview of metabolic reprogramming in urological tumors and to identify potential metabolic targets for innovative therapies.

代谢重编程是癌症的一个标志,使肿瘤细胞能够维持生长,逃避免疫监视,并抵抗治疗。泌尿系统肿瘤,包括前列腺癌、膀胱癌和肾癌,由于其独特的肿瘤微环境和致癌途径,表现出不同的代谢表型。这篇综述探讨了泌尿系统癌症肿瘤代谢的新前景,强调了关键的代谢途径,如糖酵解、脂质代谢、氨基酸代谢和氧化还原平衡。我们讨论了这些途径是如何与肿瘤进展、治疗抵抗和免疫逃避错综复杂地联系在一起的。此外,我们研究了针对代谢脆弱性的新治疗策略,包括代谢酶抑制剂、合成致死性方法和代谢调节来增强免疫治疗。通过整合多组学技术和临床前模型的进展,我们提出了一个将代谢研究转化为临床应用的框架。本综述旨在提供泌尿系统肿瘤代谢重编程的全面概述,并确定创新治疗的潜在代谢靶点。
{"title":"Metabolic Reprogramming in Urological Tumors: New Perspectives from Tumor Metabolic Phenotypes to Therapeutic Targets.","authors":"Zhuohang Li, Lin Yang, Weijia Li, Wenxue Huang, Cunzhen Ma, Boyuan Sun, Xunguo Yang, Haoxiang Xu, Zhibiao Li, Jie Zhao, Bisheng Cheng, Peng Wu","doi":"10.7150/ijbs.123647","DOIUrl":"10.7150/ijbs.123647","url":null,"abstract":"<p><p>Metabolic reprogramming is a hallmark of cancer, enabling tumor cells to sustain growth, evade immune surveillance, and resist therapy. Urological tumors, including prostate, bladder, and renal cancers, exhibit distinct metabolic phenotypes driven by their unique tumor microenvironments and oncogenic pathways. This review explores the emerging landscape of tumor metabolism in urological cancers, highlighting key metabolic pathways such as glycolysis, lipid metabolism, amino acid metabolism, and redox balance. We discuss how these pathways are intricately linked to tumor progression, therapeutic resistance, and immune evasion. Furthermore, we examine novel therapeutic strategies targeting metabolic vulnerabilities, including metabolic enzyme inhibitors, synthetic lethality approaches, and metabolic modulation to enhance immunotherapy. By integrating advances in multi-omics technologies and preclinical models, we propose a framework for translating metabolic research into clinical applications. This review aims to provide a comprehensive overview of metabolic reprogramming in urological tumors and to identify potential metabolic targets for innovative therapies.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":"21 15","pages":"6926-6951"},"PeriodicalIF":10.0,"publicationDate":"2025-10-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12631189/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145587248","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}
引用次数: 0
TGF-β1/SMAD3-mediated Non-canonical Hedgehog Signaling Promotes Pancreatic Stellate Cell Activation and Fibrosis in Chronic Pancreatitis. TGF-β1/ smad3介导的非规范Hedgehog信号通路促进慢性胰腺炎胰腺星状细胞活化和纤维化
IF 1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-10-27 eCollection Date: 2025-01-01 DOI: 10.7150/ijbs.108149
Linrui Peng, Yuchen Hu, Xiaoying Zhang, Chunlu Tan, Chan Yang, Tingting Liu, Pawel E Ferdek, Shufen Yin, Liu Wang, Wei Huang, Yuwei Zhang

Excessive Hedgehog (Hh) signaling activity contributes to fibrosis in multiple organs. However, its role in pancreatic stellate cell (PSC) activation and fibrosis development during chronic pancreatitis (CP) remains elusive. We show that GLI2 is one of the top-ranked effectors in the pancreas of CP patients and is highly expressed in activated PSCs. PSC-specific deletion of Gli2, but not Smo, significantly reduces fibrosis and the severity of the mouse CP, indicating that GLI2 in PSCs can be driven by non-canonical fashion during CP. In culture-activated primary PSCs, early nuclear translocation and increased GLI2 expression are observed promptly following in vitro culture. Whereas GLI2 inhibition reduces PSC activation, SMO inhibition dose not consistently affect changes in GLI2 levels or PSC activation. TGF-β1 promotes GLI2 activation and expression, while these processes and resultant PSC activation are reversed by TGF-β1/SMAD3 inhibition. Altogether, these findings demonstrate the activation of the non-canonical Hh pathway in PSCs during CP and highlight that GLI2 represents a promising therapeutic target for CP.

过多的刺猬(Hh)信号活动有助于多器官纤维化。然而,其在慢性胰腺炎(CP)中胰腺星状细胞(PSC)激活和纤维化发展中的作用尚不清楚。我们发现GLI2是CP患者胰腺中排名第一的效应器之一,在活化的PSCs中高度表达。psc特异性缺失Gli2,而非Smo,显著降低了纤维化和小鼠CP的严重程度,表明psc中的Gli2可以在CP期间通过非规范方式驱动。在培养激活的原代psc中,在体外培养后立即观察到早期核易位和Gli2表达增加。虽然GLI2抑制会降低PSC的激活,但SMO抑制并不会持续影响GLI2水平或PSC激活的变化。TGF-β1促进GLI2的激活和表达,而TGF-β1/SMAD3抑制可逆转这些过程和由此产生的PSC激活。总之,这些发现证明了CP期间PSCs中非规范Hh通路的激活,并强调GLI2代表了CP的一个有希望的治疗靶点。
{"title":"TGF-β1/SMAD3-mediated Non-canonical Hedgehog Signaling Promotes Pancreatic Stellate Cell Activation and Fibrosis in Chronic Pancreatitis.","authors":"Linrui Peng, Yuchen Hu, Xiaoying Zhang, Chunlu Tan, Chan Yang, Tingting Liu, Pawel E Ferdek, Shufen Yin, Liu Wang, Wei Huang, Yuwei Zhang","doi":"10.7150/ijbs.108149","DOIUrl":"10.7150/ijbs.108149","url":null,"abstract":"<p><p>Excessive Hedgehog (Hh) signaling activity contributes to fibrosis in multiple organs. However, its role in pancreatic stellate cell (PSC) activation and fibrosis development during chronic pancreatitis (CP) remains elusive. We show that GLI2 is one of the top-ranked effectors in the pancreas of CP patients and is highly expressed in activated PSCs. PSC-specific deletion of <i>Gli2</i>, but not <i>Smo</i>, significantly reduces fibrosis and the severity of the mouse CP, indicating that GLI2 in PSCs can be driven by non-canonical fashion during CP. In culture-activated primary PSCs, early nuclear translocation and increased GLI2 expression are observed promptly following in vitro culture. Whereas GLI2 inhibition reduces PSC activation, SMO inhibition dose not consistently affect changes in GLI2 levels or PSC activation. TGF-β1 promotes GLI2 activation and expression, while these processes and resultant PSC activation are reversed by TGF-β1/SMAD3 inhibition. Altogether, these findings demonstrate the activation of the non-canonical Hh pathway in PSCs during CP and highlight that GLI2 represents a promising therapeutic target for CP.</p>","PeriodicalId":13762,"journal":{"name":"International Journal of Biological Sciences","volume":"21 15","pages":"6978-6996"},"PeriodicalIF":10.0,"publicationDate":"2025-10-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12631176/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145587331","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}
引用次数: 0
期刊
International Journal of Biological Sciences
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1