Pub Date : 2025-12-21DOI: 10.1016/j.bbadis.2025.168145
Jiancheng Mou , Hongchao Tang , Xiaoge Hu , Zhuotao Yang , Haotian Su , Da Qian , Chenhong Li , Haotian Liu , Zhihao Ye , Mingxing Xu , Shuyan Liu , Qinghui Zheng , Xiaozhen Liu , Xin Zeng , Qiuran Xu , Xuli Meng
Background
Breast cancer (BC) remains one of the major threats to women's health in the 21st century, due to its high incidence and mortality rates. Ubiquitin-conjugating enzymes, as members of the ubiquitin-proteasome system, are responsible for numerous cellular physiological processes. However, ubiquitin-conjugating enzymes may also play unexpected roles in other physiological activities, such as phosphorylation, lactylation, and even methylation. The physiological function of the ubiquitin-conjugating E2 enzyme UBE2K in BC remains unknown. As a result, we looked into UBE2K's physiological role in the malignant development of BC.
Methods
A combination of RT-qPCR, Transwell migration assays, Western blotting, and CCK-8 analysis was employed to confirm the upregulation of UBE2K in BC cells and to assess its role in promoting cell proliferation and migration. Furthermore, using chromatin immunoprecipitation (ChIP) and dual-luciferase reporter assays, we identified and validated CREB1 as a transcription factor for UBE2K for the first time.
Results
We discovered that UBE2K regulates the physiological processes of BC cells via the STUB1/PKA/CREB1/p-CREB1 axis. Moreover, functional rescue experiments ultimately displayed that UBE2K promotes the malignant progression of BC cells by via STUB1/PKA/CREB1/p-CREB1 axis.
Conclusions
In conclusion, the UBE2K/CREB1 positive feedback loop promotes the development of BC, indicating that UBE2K could be a viable therapeutic target for anti-BC.
{"title":"UBE2K promotes breast cancer growth by ubiquitinating and degrading STUB1 to regulate the PKA/CREB1 signaling pathway, forming a feedback loop","authors":"Jiancheng Mou , Hongchao Tang , Xiaoge Hu , Zhuotao Yang , Haotian Su , Da Qian , Chenhong Li , Haotian Liu , Zhihao Ye , Mingxing Xu , Shuyan Liu , Qinghui Zheng , Xiaozhen Liu , Xin Zeng , Qiuran Xu , Xuli Meng","doi":"10.1016/j.bbadis.2025.168145","DOIUrl":"10.1016/j.bbadis.2025.168145","url":null,"abstract":"<div><h3>Background</h3><div>Breast cancer (BC) remains one of the major threats to women's health in the 21st century, due to its high incidence and mortality rates. Ubiquitin-conjugating enzymes, as members of the ubiquitin-proteasome system, are responsible for numerous cellular physiological processes. However, ubiquitin-conjugating enzymes may also play unexpected roles in other physiological activities, such as phosphorylation, lactylation, and even methylation. The physiological function of the ubiquitin-conjugating E2 enzyme UBE2K in BC remains unknown. As a result, we looked into UBE2K's physiological role in the malignant development of BC.</div></div><div><h3>Methods</h3><div>A combination of RT-qPCR, Transwell migration assays, Western blotting, and CCK-8 analysis was employed to confirm the upregulation of UBE2K in BC cells and to assess its role in promoting cell proliferation and migration. Furthermore, using chromatin immunoprecipitation (ChIP) and dual-luciferase reporter assays, we identified and validated CREB1 as a transcription factor for UBE2K for the first time.</div></div><div><h3>Results</h3><div>We discovered that UBE2K regulates the physiological processes of BC cells via the STUB1/PKA/CREB1/p-CREB1 axis. Moreover, functional rescue experiments ultimately displayed that UBE2K promotes the malignant progression of BC cells by via STUB1/PKA/CREB1/p-CREB1 axis.</div></div><div><h3>Conclusions</h3><div>In conclusion, the UBE2K/CREB1 positive feedback loop promotes the development of BC, indicating that UBE2K could be a viable therapeutic target for anti-BC.</div></div>","PeriodicalId":8821,"journal":{"name":"Biochimica et biophysica acta. Molecular basis of disease","volume":"1872 3","pages":"Article 168145"},"PeriodicalIF":4.2,"publicationDate":"2025-12-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145822347","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 : 2025-12-21DOI: 10.1016/j.bbadis.2025.168143
Sally H. Fayad , Alaa Mohamed Salah , Ayad A. Jaffa , Nadine Darwiche
Acute myeloid leukemia (AML) is one of the most frequent hematological malignancies. It is a complex and aggressive disease of undifferentiated hematopoietic progenitor cells, with high rates of relapse and drug resistance, posing significant clinical challenges. Emerging evidence underscores the critical role of inflammation in AML progression, with inflammasomes—key regulators of the inflammatory response—emerging as pivotal players in disease pathogenesis. Among these, the NOD-, LRR-, and pyrin domain-containing 3 (NLRP3) inflammasome has gained particular attention due to its involvement in promoting leukemogenesis and chemoresistance. While inflammasomes have been previously studied in various cancer types, no comprehensive review to date has specifically addressed the role of the NLRP3 inflammasome in AML and its potential targeted therapy. Focusing on the NLRP3 inflammasome, we explore its contribution to AML pathogenesis and its therapeutic potential. We highlight preclinical natural and synthetic compounds, alongside clinical-stage and clinically approved drugs that target the NLRP3 inflammasome signalling pathway and components. We address the challenges and limitations of these promising compounds and drugs, while highlighting recent advancements such as the synthesis of novel and derivatives designed to improve their efficacy. Finally, we discuss future research directions aimed at deepening our understanding of the NLRP3 inflammasome and identifying novel and improved therapeutic outcomes against AML.
{"title":"Targeting the NLRP3 inflammasome signalling in acute myeloid leukemia: Mechanisms, therapeutics, and future directions","authors":"Sally H. Fayad , Alaa Mohamed Salah , Ayad A. Jaffa , Nadine Darwiche","doi":"10.1016/j.bbadis.2025.168143","DOIUrl":"10.1016/j.bbadis.2025.168143","url":null,"abstract":"<div><div>Acute myeloid leukemia (AML) is one of the most frequent hematological malignancies. It is a complex and aggressive disease of undifferentiated hematopoietic progenitor cells, with high rates of relapse and drug resistance, posing significant clinical challenges. Emerging evidence underscores the critical role of inflammation in AML progression, with inflammasomes—key regulators of the inflammatory response—emerging as pivotal players in disease pathogenesis. Among these, the NOD-, LRR-, and pyrin domain-containing 3 (NLRP3) inflammasome has gained particular attention due to its involvement in promoting leukemogenesis and chemoresistance. While inflammasomes have been previously studied in various cancer types, no comprehensive review to date has specifically addressed the role of the NLRP3 inflammasome in AML and its potential targeted therapy. Focusing on the NLRP3 inflammasome, we explore its contribution to AML pathogenesis and its therapeutic potential. We highlight preclinical natural and synthetic compounds, alongside clinical-stage and clinically approved drugs that target the NLRP3 inflammasome signalling pathway and components. We address the challenges and limitations of these promising compounds and drugs, while highlighting recent advancements such as the synthesis of novel and derivatives designed to improve their efficacy. Finally, we discuss future research directions aimed at deepening our understanding of the NLRP3 inflammasome and identifying novel and improved therapeutic outcomes against AML.</div></div>","PeriodicalId":8821,"journal":{"name":"Biochimica et biophysica acta. Molecular basis of disease","volume":"1872 3","pages":"Article 168143"},"PeriodicalIF":4.2,"publicationDate":"2025-12-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145822341","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 : 2025-12-18DOI: 10.1016/j.bbadis.2025.168140
Yanan Zhang , Lei Li , Gaiqin Li , Junyuan Zhu , Zexue Qi , Chunqing Zhang
Purpose
We aimed to investigate the role and possible mechanism of action of hexokinase domain containing 1 (HKDC1) in liver cirrhosis.
Methods
Hepatic stellate cell (HSC) activation model was established in LX-2 cells by transforming growth factor (TGF)-βl stimulation. A mouse model of hepatic fibrosis was established using carbon tetrachloride (CCl4) stimulation. HKDC1 expression was assessed using qRT-PCR, western blotting, and immunofluorescence. Hematoxylin and eosin (H&E) and Masson staining were used to assess liver injury and fibrosis. Changes in fibrosis markers were assessed using qRT-PCR, western blotting, and immunohistochemical staining. The effect of HKDC1 on glycolysis was evaluated by measuring the levels of extracellular acidification rate (ECAR), oxygen consumption rate (OCR), lactate, and related proteins.
Results
In liver cirrhosis tissues and activated HSCs, HKDC1 expression was upregulated. CCl4-induced liver injury and fibrosis were inhibited by silencing of Hkdc1 in mice, as evidenced by the decrease of aspartate transaminase (AST), alanine transaminase (ALT), Collagen I, α-SMA, TGF-β1, and TIMP-1. Under TGF-β1 treatment, silencing of HKDC1 inhibited HSC activation and glycolysis, as evidenced by the reduce of Collagen I, α-SMA, TIMP-1, ECAR, lactate, HK2, LDHA, PKM2 and the increase of OCR. Mechanistically, silencing of HKDC1 reduced the levels of ORMDL3 and H3K18la proteins, and HKDC1 increased histone lactylation of the promoter of ORMDL3. ORMDL3 overexpression and lactate eliminated the effects of HKDC1 silencing on LX-2 cell activation.
Conclusion
HKDC1 silencing alleviates liver fibrosis and HSC activation by regulating glycolysis and decreasing histone lactylation of the promoter of ORMDL3.
{"title":"HKDC1 promotes the H3K18 lactylation of the promoter of ORMDL3 to induce the activation of hepatic stellate cells in liver cirrhosis","authors":"Yanan Zhang , Lei Li , Gaiqin Li , Junyuan Zhu , Zexue Qi , Chunqing Zhang","doi":"10.1016/j.bbadis.2025.168140","DOIUrl":"10.1016/j.bbadis.2025.168140","url":null,"abstract":"<div><h3>Purpose</h3><div>We aimed to investigate the role and possible mechanism of action of hexokinase domain containing 1 (HKDC1) in liver cirrhosis.</div></div><div><h3>Methods</h3><div>Hepatic stellate cell (HSC) activation model was established in LX-2 cells by transforming growth factor (TGF)-βl stimulation. A mouse model of hepatic fibrosis was established using carbon tetrachloride (CCl<sub>4</sub>) stimulation. HKDC1 expression was assessed using qRT-PCR, western blotting, and immunofluorescence. Hematoxylin and eosin (H&E) and Masson staining were used to assess liver injury and fibrosis. Changes in fibrosis markers were assessed using qRT-PCR, western blotting, and immunohistochemical staining. The effect of HKDC1 on glycolysis was evaluated by measuring the levels of extracellular acidification rate (ECAR), oxygen consumption rate (OCR), lactate, and related proteins.</div></div><div><h3>Results</h3><div>In liver cirrhosis tissues and activated HSCs, HKDC1 expression was upregulated. CCl<sub>4</sub>-induced liver injury and fibrosis were inhibited by silencing of <em>Hkdc1</em> in mice, as evidenced by the decrease of aspartate transaminase (AST), alanine transaminase (ALT), Collagen I, α-SMA, TGF-β1, and TIMP-1. Under TGF-β1 treatment, silencing of <em>HKDC1</em> inhibited HSC activation and glycolysis, as evidenced by the reduce of Collagen I, α-SMA, TIMP-1, ECAR, lactate, HK2, LDHA, PKM2 and the increase of OCR. Mechanistically, silencing of <em>HKDC1</em> reduced the levels of ORMDL3 and H3K18la proteins, and HKDC1 increased histone lactylation of the promoter of <em>ORMDL3</em>. <em>ORMDL3</em> overexpression and lactate eliminated the effects of <em>HKDC1</em> silencing on LX-2 cell activation.</div></div><div><h3>Conclusion</h3><div><em>HKDC1</em> silencing alleviates liver fibrosis and HSC activation by regulating glycolysis and decreasing histone lactylation of the promoter of <em>ORMDL3</em>.</div></div>","PeriodicalId":8821,"journal":{"name":"Biochimica et biophysica acta. Molecular basis of disease","volume":"1872 3","pages":"Article 168140"},"PeriodicalIF":4.2,"publicationDate":"2025-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145786500","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 : 2025-12-17DOI: 10.1016/j.bbadis.2025.168142
Shobini Jayaraman , Kyeongseo Choi , Antonio Pérez , Inka Miñambres , Jose Luis Sánchez-Quesada , Olga Gursky
Low-density lipoprotein (LDL) entrapment in the extracellular matrix of the arterial intima is an established early trigger of atherosclerosis. Though much is known about LDL binding to arterial proteoglycans, the binding to other matrix components remains underexplored. This study focuses on LDL interactions with type-I collagen (Col-I), a major extracellular matrix protein found in atherosclerotic lesions. Human normolipidemic LDL, which has been oxidized or lipolyzed in vitro, and naturally occurring plasma LDL subclasses differing in size and charge are explored, along with LDL from patients with type-2 diabetes or obesity before and after treatment. To understand how these lipoproteins interact with Col-I, we quantify their binding to collagen-coated microwell plates using enzyme-linked immunosorbent assay. The mechanistic underpinnings are probed by a multipronged approach using the methods of chromatography, spectroscopy, microscopy and others. The results reveal that intact LDL binds Col-I in a cooperative dose-dependent manner. The binding is synergistic with pro-atherogenic LDL modifications including aggregation, oxidation and lipolysis. The pro-atherogenic subclasses of small dense LDL and electronegative LDL show enhanced Col-I binding; conversely, Col-I binding induces LDL remodeling to generate smaller and more electronegative particles. This synergy suggests a potential causal role of LDL–Col-I interactions in atherogenesis. Moreover, our results suggest that LDL binding to Col-I provides a biomarker and a sensitive treatment readout for the risk of atherosclerosis in patients with metabolic diseases such as diabetes and obesity.
{"title":"Binding of human low-density lipoproteins to type-I collagen is synergistic with the pro-atherogenic lipoprotein remodeling","authors":"Shobini Jayaraman , Kyeongseo Choi , Antonio Pérez , Inka Miñambres , Jose Luis Sánchez-Quesada , Olga Gursky","doi":"10.1016/j.bbadis.2025.168142","DOIUrl":"10.1016/j.bbadis.2025.168142","url":null,"abstract":"<div><div>Low-density lipoprotein (LDL) entrapment in the extracellular matrix of the arterial intima is an established early trigger of atherosclerosis. Though much is known about LDL binding to arterial proteoglycans, the binding to other matrix components remains underexplored. This study focuses on LDL interactions with type-I collagen (Col-I), a major extracellular matrix protein found in atherosclerotic lesions. Human normolipidemic LDL, which has been oxidized or lipolyzed in vitro, and naturally occurring plasma LDL subclasses differing in size and charge are explored, along with LDL from patients with type-2 diabetes or obesity before and after treatment. To understand how these lipoproteins interact with Col-I, we quantify their binding to collagen-coated microwell plates using enzyme-linked immunosorbent assay. The mechanistic underpinnings are probed by a multipronged approach using the methods of chromatography, spectroscopy, microscopy and others. The results reveal that intact LDL binds Col-I in a cooperative dose-dependent manner. The binding is synergistic with pro-atherogenic LDL modifications including aggregation, oxidation and lipolysis. The pro-atherogenic subclasses of small dense LDL and electronegative LDL show enhanced Col-I binding; conversely, Col-I binding induces LDL remodeling to generate smaller and more electronegative particles. This synergy suggests a potential causal role of LDL–Col-I interactions in atherogenesis. Moreover, our results suggest that LDL binding to Col-I provides a biomarker and a sensitive treatment readout for the risk of atherosclerosis in patients with metabolic diseases such as diabetes and obesity.</div></div>","PeriodicalId":8821,"journal":{"name":"Biochimica et biophysica acta. Molecular basis of disease","volume":"1872 3","pages":"Article 168142"},"PeriodicalIF":4.2,"publicationDate":"2025-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145795352","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 : 2025-12-17DOI: 10.1016/j.bbadis.2025.168144
Benjamin R. Pryce , Haiming L. Kerr
Cancer cachexia is characterized by a significant loss in body weight due to the wasting of skeletal muscle and adipose tissue. Systemic inflammation has long been associated with cachexia, with various tumor secreted factors shown to correlate with as well as cause tissue wasting. In addition to systemic inflammation, it has become increasingly appreciated that inflammation occurs in specific tissues in cancer cachexia, with tissues such as muscle, adipose, liver and brain being affected. While several studies have shown that this local tissue inflammation contributes to cachexia, there is evidence that some aspects of the inflammatory response may play a protective role to mitigate tissue wasting. Here, we will review the findings on local tissue inflammation in cachexia, comparing the impacts of such inflammation on tissue wasting and cachexia progression overall. Furthermore, we discuss the methods used to mitigate inflammation in various tissues and highlight the outcomes on the cachectic phenotype. Collectively, understanding how inflammation contributes to cachexia in each tissue will ultimately influence how therapies can be designed to treat cachexia while minimizing possible adverse side effects.
{"title":"The effects of tissue inflammation on cancer cachexia","authors":"Benjamin R. Pryce , Haiming L. Kerr","doi":"10.1016/j.bbadis.2025.168144","DOIUrl":"10.1016/j.bbadis.2025.168144","url":null,"abstract":"<div><div>Cancer cachexia is characterized by a significant loss in body weight due to the wasting of skeletal muscle and adipose tissue. Systemic inflammation has long been associated with cachexia, with various tumor secreted factors shown to correlate with as well as cause tissue wasting. In addition to systemic inflammation, it has become increasingly appreciated that inflammation occurs in specific tissues in cancer cachexia, with tissues such as muscle, adipose, liver and brain being affected. While several studies have shown that this local tissue inflammation contributes to cachexia, there is evidence that some aspects of the inflammatory response may play a protective role to mitigate tissue wasting. Here, we will review the findings on local tissue inflammation in cachexia, comparing the impacts of such inflammation on tissue wasting and cachexia progression overall. Furthermore, we discuss the methods used to mitigate inflammation in various tissues and highlight the outcomes on the cachectic phenotype. Collectively, understanding how inflammation contributes to cachexia in each tissue will ultimately influence how therapies can be designed to treat cachexia while minimizing possible adverse side effects.</div></div>","PeriodicalId":8821,"journal":{"name":"Biochimica et biophysica acta. Molecular basis of disease","volume":"1872 3","pages":"Article 168144"},"PeriodicalIF":4.2,"publicationDate":"2025-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145795389","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 : 2025-12-16DOI: 10.1016/j.bbadis.2025.168134
Yuling Xie , Xinfan Lin , Xu Han , Xingfeng Chen , Qingsong Wu , Debin Jiang , Linfeng Xie
Background
The development of aortic dissection (AD) is closely associated with extracellular matrix degradation and the apoptosis of vascular smooth muscle cells (VSMCs). Antioxidant-1 (ATOX1), a copper-binding protein, the precise mechanisms by which it contributes to extracellular matrix (ECM) degradation, VSMC apoptosis, and the onset of AD remain to be further elucidated.
Methods and results
Through high-throughput sequencing, we identified a significant increase in the expression of ATOX1 in patients with AD. Further validation using tissue staining, RT-PCR and Western blot revealed that ATOX1 expression was elevated in AD patients, AD mouse models, and in vitro human aortic vascular smooth muscle cells (HAVSMCs) induced by Angiotensin II (AngII). In vitro experiments showed that silencing ATOX1 or pharmacologically inhibiting ATOX1 with DC_AC50 significantly reduced copper ion expression and the secretion of matrix metalloproteinases (MMPs), while alleviating cell apoptosis in HAVSMCs. Targeted knockdown of ATOX1 in smooth muscle cells using adeno-associated virus vector 9 (AAV9) or pharmacological inhibition of ATOX1 effectively slowed the progression of AD in a β-aminopropionitrile (BAPN)-induced mouse model. Additionally, ATOX1 expression is directly regulated by miR-133b, which was found to be significantly downregulated in the serum and aortic tissues of AD patients, exhibiting an inverse correlation with ATOX1 upregulation in AD. MiR-133b mimic successfully reversed the effects of ATOX1-induced MMPs secretion and apoptosis in HAVSMCs. Lastly, overexpression of miR-133b through AAV9 significantly attenuated the progression of BAPN-induced AD in mice.
Conclusions
Our study suggests that inhibiting ATOX1 may reduce ECM degradation and cell apoptosis, thereby slowing the progression of AD, and highlights ATOX1 inhibition as a potential new strategy for AD treatment.
{"title":"ATOX1-driven ECM degradation and vascular smooth muscle cell apoptosis accelerate aortic dissection progression","authors":"Yuling Xie , Xinfan Lin , Xu Han , Xingfeng Chen , Qingsong Wu , Debin Jiang , Linfeng Xie","doi":"10.1016/j.bbadis.2025.168134","DOIUrl":"10.1016/j.bbadis.2025.168134","url":null,"abstract":"<div><h3>Background</h3><div>The development of aortic dissection (AD) is closely associated with extracellular matrix degradation and the apoptosis of vascular smooth muscle cells (VSMCs). Antioxidant-1 (ATOX1), a copper-binding protein, the precise mechanisms by which it contributes to extracellular matrix (ECM) degradation, VSMC apoptosis, and the onset of AD remain to be further elucidated.</div></div><div><h3>Methods and results</h3><div>Through high-throughput sequencing, we identified a significant increase in the expression of ATOX1 in patients with AD. Further validation using tissue staining, RT-PCR and Western blot revealed that ATOX1 expression was elevated in AD patients, AD mouse models, and in vitro human aortic vascular smooth muscle cells (HAVSMCs) induced by Angiotensin II (AngII). In vitro experiments showed that silencing ATOX1 or pharmacologically inhibiting ATOX1 with DC_AC50 significantly reduced copper ion expression and the secretion of matrix metalloproteinases (MMPs), while alleviating cell apoptosis in HAVSMCs. Targeted knockdown of ATOX1 in smooth muscle cells using adeno-associated virus vector 9 (AAV9) or pharmacological inhibition of ATOX1 effectively slowed the progression of AD in a β-aminopropionitrile (BAPN)-induced mouse model. Additionally, ATOX1 expression is directly regulated by miR-133b, which was found to be significantly downregulated in the serum and aortic tissues of AD patients, exhibiting an inverse correlation with ATOX1 upregulation in AD. MiR-133b mimic successfully reversed the effects of ATOX1-induced MMPs secretion and apoptosis in HAVSMCs. Lastly, overexpression of miR-133b through AAV9 significantly attenuated the progression of BAPN-induced AD in mice.</div></div><div><h3>Conclusions</h3><div>Our study suggests that inhibiting ATOX1 may reduce ECM degradation and cell apoptosis, thereby slowing the progression of AD, and highlights ATOX1 inhibition as a potential new strategy for AD treatment.</div></div>","PeriodicalId":8821,"journal":{"name":"Biochimica et biophysica acta. Molecular basis of disease","volume":"1872 3","pages":"Article 168134"},"PeriodicalIF":4.2,"publicationDate":"2025-12-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145784090","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 : 2025-12-15DOI: 10.1016/j.bbadis.2025.168139
Weibo Huang , Chenghao Zhou , Rong Wu , Kangcheng Zhao , Jianfeng Wu , Dianwen Song
Objective
Type 2 diabetes (T2D) is associated with osteoporosis. Although chronic inflammation and immune dysregulation are implicated in T2D-induced bone loss, the specific roles of immune cells remain poorly understood. This study aimed to investigate how neutrophils in T2D contribute to osteoporosis and to identify the underlying molecular mechanisms and potential therapeutic targets.
Methods
We utilized a combination of in vivo and in vitro approaches, including T2D and control mouse models, primary cell cultures, and publicly available single-cell RNA sequencing data from the GEO database. Osteoporosis was assessed through TRAP staining, micro-CT imaging, and quantitative PCR. Molecular interactions were examined using Western blotting, chromatin immunoprecipitation followed by qPCR, and flow cytometry. Additionally, virtual screening was employed to identify potential inhibitors of thrombospondin-1(THBS1).
Results
Neutrophils isolated from T2D mice promoted osteoclast activity and bone loss when transferred into wild-type mice, as evidenced by increased TRAP-positive cells and deteriorated bone microarchitecture. We identified THBS1, a glycoprotein secreted at higher levels by T2D neutrophils, as a mediator of osteoclast differentiation. THBS1 engaged the CD36 receptor on macrophages, activating PPARγ, which transcriptionally upregulates POU2F2—a transcription factor that enhances osteoclastogenesis via c-FOS induction. Genetic ablation of THBS1 attenuated osteoclast formation and bone loss in T2D mice. Virtual screening identified nasunin as a potent THBS1 inhibitor. Treatment with nasunin suppressed the CD36–PPARγ–POU2F2–c-FOS axis, reduced osteoclast differentiation in vitro, and ameliorated T2D-induced osteoporosis in vivo.
Conclusion
Our findings reveal a novel pathway through which neutrophil-derived THBS1 exacerbates diabetic osteoporosis by promoting osteoclastogenesis via CD36–PPARγ–POU2F2 signaling.
{"title":"Neutrophil-derived thrombospondin-1 (THBS1) drives type 2 diabetes-induced osteoporosis via CD36-PPARγ-POU2F2 signaling","authors":"Weibo Huang , Chenghao Zhou , Rong Wu , Kangcheng Zhao , Jianfeng Wu , Dianwen Song","doi":"10.1016/j.bbadis.2025.168139","DOIUrl":"10.1016/j.bbadis.2025.168139","url":null,"abstract":"<div><h3>Objective</h3><div>Type 2 diabetes (T2D) is associated with osteoporosis. Although chronic inflammation and immune dysregulation are implicated in T2D-induced bone loss, the specific roles of immune cells remain poorly understood. This study aimed to investigate how neutrophils in T2D contribute to osteoporosis and to identify the underlying molecular mechanisms and potential therapeutic targets.</div></div><div><h3>Methods</h3><div>We utilized a combination of in vivo and in vitro approaches, including T2D and control mouse models, primary cell cultures, and publicly available single-cell RNA sequencing data from the GEO database. Osteoporosis was assessed through TRAP staining, micro-CT imaging, and quantitative PCR. Molecular interactions were examined using Western blotting, chromatin immunoprecipitation followed by qPCR, and flow cytometry. Additionally, virtual screening was employed to identify potential inhibitors of thrombospondin-1(THBS1).</div></div><div><h3>Results</h3><div>Neutrophils isolated from T2D mice promoted osteoclast activity and bone loss when transferred into wild-type mice, as evidenced by increased TRAP-positive cells and deteriorated bone microarchitecture. We identified THBS1, a glycoprotein secreted at higher levels by T2D neutrophils, as a mediator of osteoclast differentiation. THBS1 engaged the CD36 receptor on macrophages, activating PPARγ, which transcriptionally upregulates POU2F2—a transcription factor that enhances osteoclastogenesis via c-FOS induction. Genetic ablation of THBS1 attenuated osteoclast formation and bone loss in T2D mice. Virtual screening identified nasunin as a potent THBS1 inhibitor. Treatment with nasunin suppressed the CD36–PPARγ–POU2F2–c-FOS axis, reduced osteoclast differentiation in vitro, and ameliorated T2D-induced osteoporosis in vivo.</div></div><div><h3>Conclusion</h3><div>Our findings reveal a novel pathway through which neutrophil-derived THBS1 exacerbates diabetic osteoporosis by promoting osteoclastogenesis via CD36–PPARγ–POU2F2 signaling.</div></div>","PeriodicalId":8821,"journal":{"name":"Biochimica et biophysica acta. Molecular basis of disease","volume":"1872 3","pages":"Article 168139"},"PeriodicalIF":4.2,"publicationDate":"2025-12-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145776762","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 : 2025-12-15DOI: 10.1016/j.bbadis.2025.168131
Qianqian Han , Yutong Zou , Qing Yang , Enrong Ran , Ziyao Li , Fang Liu
CD4+ T cells play a critical role in diabetic nephropathy (DN) progression. High-glucose (HG) conditions induce small extracellular vesicle (sEV) cargo loading disorders, but whether CD4+ T cells mediate renal injury via sEVs remains unclear. In vitro, a transformed C3H mouse kidney-1 (TCMK-1)/CD4+ T-cell coculture model revealed that high-glucose (HG) conditions increased CD4+ T-cell-induced lactate dehydrogenase release, reactive oxygen species production, apoptosis of TCMK-1 cells, and mitochondrial dysfunction, as confirmed by enzyme-linked immunosorbent assays, flow cytometry, Western blotting (WB), and transmission electron microscopy (TEM). Notably, compared with those under normal glucose conditions, CD4+ T cells under HG conditions exacerbated these effects more significantly. In vivo, CD4+ T cells were isolated from the blood of C57BL/6J (control) mice and db/db (experimental) mice using magnetic beads, and their sEVs were extracted from the culture medium. These sEVs were injected into corresponding C57BL/6J or db/db mice, with saline-injected mice used as blank controls. Compared with control mice, mice injected with db/db mouse CD4+ T-cell-derived sEVs (both C57BL/6J and db/db recipients) exhibited significant increases in serum creatinine levels and proteinuria, as well as more severe renal pathological injury. TEM, flow cytometry and WB analyses revealed disordered mitochondrial dynamics in renal tubular epithelial cells (RTECs), marked by reduced membrane potential and imbalanced Drp1/Mfn1/Mfn2 expression. Collectively, these findings indicate that CD4+ T cells exacerbate renal injury and disrupt mitochondrial homeostasis in RTECs by releasing sEVs under HG conditions, highlighting the CD4+ T-cell-sEV axis as a potential diagnostic and therapeutic target for DN.
{"title":"CD4+ T-cell-derived small extracellular vesicles induce the apoptosis of renal tubular epithelial cells in diabetic nephropathy by regulating mitochondrial dynamics","authors":"Qianqian Han , Yutong Zou , Qing Yang , Enrong Ran , Ziyao Li , Fang Liu","doi":"10.1016/j.bbadis.2025.168131","DOIUrl":"10.1016/j.bbadis.2025.168131","url":null,"abstract":"<div><div>CD4+ T cells play a critical role in diabetic nephropathy (DN) progression. High-glucose (HG) conditions induce small extracellular vesicle (sEV) cargo loading disorders, but whether CD4+ T cells mediate renal injury via sEVs remains unclear. In vitro, a transformed C3H mouse kidney-1 (TCMK-1)/CD4+ T-cell coculture model revealed that high-glucose (HG) conditions increased CD4+ T-cell-induced lactate dehydrogenase release, reactive oxygen species production, apoptosis of TCMK-1 cells, and mitochondrial dysfunction, as confirmed by enzyme-linked immunosorbent assays, flow cytometry, Western blotting (WB), and transmission electron microscopy (TEM). Notably, compared with those under normal glucose conditions, CD4+ T cells under HG conditions exacerbated these effects more significantly. In vivo, CD4+ T cells were isolated from the blood of C57BL/6J (control) mice and db/db (experimental) mice using magnetic beads, and their sEVs were extracted from the culture medium. These sEVs were injected into corresponding C57BL/6J or db/db mice, with saline-injected mice used as blank controls. Compared with control mice, mice injected with db/db mouse CD4+ T-cell-derived sEVs (both C57BL/6J and db/db recipients) exhibited significant increases in serum creatinine levels and proteinuria, as well as more severe renal pathological injury. TEM, flow cytometry and WB analyses revealed disordered mitochondrial dynamics in renal tubular epithelial cells (RTECs), marked by reduced membrane potential and imbalanced Drp1/Mfn1/Mfn2 expression. Collectively, these findings indicate that CD4+ T cells exacerbate renal injury and disrupt mitochondrial homeostasis in RTECs by releasing sEVs under HG conditions, highlighting the CD4+ T-cell-sEV axis as a potential diagnostic and therapeutic target for DN.</div></div>","PeriodicalId":8821,"journal":{"name":"Biochimica et biophysica acta. Molecular basis of disease","volume":"1872 3","pages":"Article 168131"},"PeriodicalIF":4.2,"publicationDate":"2025-12-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145770300","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 : 2025-12-15DOI: 10.1016/j.bbadis.2025.168138
Wen Fu , Qiaoliang Wu , Yanru Yao , Yingsen Sun , Huafang Wang , Jing Jie , Xiaoyan Zheng , Lina Wang , Xiaoge Hu , Xiangmin Tong , Qiuran Xu
Background
Ubiquitin-specific peptidase 40(USP40), a member of the deubiquitinase family, regulates numerous cellular processes. Recent studies have increasingly highlighted the involvement of USP40 in cancer development. However, the precise mechanism through which USP40 influences acute myeloid leukemia (AML) progression remains poorly understood.
Methods
The expression levels of USP40 in AML tissues and cell lines were examined using Gene Expression Profiling Interactive Analysis, reverse transcription quantitative polymerase chain reaction (RT-qPCR), and western blotting (WB). The Kaplan-Meier plotter was used to evaluate the association between USP40 expression and prognosis in patients with AML. Cell Counting Kit-8, 5-ethynyl-2′-deoxyuridine incorporation, apoptosis assays, and subcutaneous tumor models in nude mice were applied to determine the effects of USP40 expression on AML cell proliferation and apoptosis in vitro and in vivo. WB, RT-qPCR, immunofluorescence assay, co-immunoprecipitation assay, and cycloheximide chase assay were conducted to explore the relationship between USP40 and c-MYC. Rescue experiments were further performed to assess the functional contribution of USP40-c-MYC axis to AML progression.
Results
USP40 was overexpressed in AML tissues and cell lines compared to normal controls and correlated with poor prognosis. USP40 accelerated AML progression by promoting proliferation and inhibiting apoptosis. Mechanistically, USP40 deubiquitinated c-MYC by selectively removing K48-linked polyubiquitin chains, thereby preventing its degradation via the ubiquitin–proteasome pathway.
Conclusion
USP40 accelerated AML progression by deubiquitinating c-MYC, highlighting the USP40-c-MYC axis as a potential therapeutic target for AML treatment.
{"title":"Ubiquitin-specific peptidase 40 promotes cellular proliferation and inhibits apoptosis through deubiquitination-mediated stabilization of c-MYC in acute myeloid leukemia","authors":"Wen Fu , Qiaoliang Wu , Yanru Yao , Yingsen Sun , Huafang Wang , Jing Jie , Xiaoyan Zheng , Lina Wang , Xiaoge Hu , Xiangmin Tong , Qiuran Xu","doi":"10.1016/j.bbadis.2025.168138","DOIUrl":"10.1016/j.bbadis.2025.168138","url":null,"abstract":"<div><h3>Background</h3><div>Ubiquitin-specific peptidase 40(USP40), a member of the deubiquitinase family, regulates numerous cellular processes. Recent studies have increasingly highlighted the involvement of USP40 in cancer development. However, the precise mechanism through which USP40 influences acute myeloid leukemia (AML) progression remains poorly understood.</div></div><div><h3>Methods</h3><div>The expression levels of USP40 in AML tissues and cell lines were examined using Gene Expression Profiling Interactive Analysis, reverse transcription quantitative polymerase chain reaction (RT-qPCR), and western blotting (WB). The Kaplan-Meier plotter was used to evaluate the association between USP40 expression and prognosis in patients with AML. Cell Counting Kit-8, 5-ethynyl-2′-deoxyuridine incorporation, apoptosis assays, and subcutaneous tumor models in nude mice were applied to determine the effects of USP40 expression on AML cell proliferation and apoptosis in vitro and in vivo. WB, RT-qPCR, immunofluorescence assay, co-immunoprecipitation assay, and cycloheximide chase assay were conducted to explore the relationship between USP40 and c-MYC. Rescue experiments were further performed to assess the functional contribution of USP40-c-MYC axis to AML progression.</div></div><div><h3>Results</h3><div>USP40 was overexpressed in AML tissues and cell lines compared to normal controls and correlated with poor prognosis. USP40 accelerated AML progression by promoting proliferation and inhibiting apoptosis. Mechanistically, USP40 deubiquitinated c-MYC by selectively removing K48-linked polyubiquitin chains, thereby preventing its degradation via the ubiquitin–proteasome pathway.</div></div><div><h3>Conclusion</h3><div>USP40 accelerated AML progression by deubiquitinating c-MYC, highlighting the USP40-c-MYC axis as a potential therapeutic target for AML treatment.</div></div>","PeriodicalId":8821,"journal":{"name":"Biochimica et biophysica acta. Molecular basis of disease","volume":"1872 3","pages":"Article 168138"},"PeriodicalIF":4.2,"publicationDate":"2025-12-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145776790","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 : 2025-12-13DOI: 10.1016/j.bbadis.2025.168132
Liman Lin , Chunle Zhao , Xiaoya Cai , Aiguo Liu , Min Xiao , Liting Chen , Dengju Li
Chemotherapy resistance represents a major challenge in relapsed and refractory acute myeloid leukemia (AML). Therefore, it is necessary to investigate the mechanisms underlying chemotherapy resistance in AML. INSR not only highly expressed in AML cells treated with chidamide, but also elevated in AML patients. Subsequent overexpression and knockdown experiments of INSR in AML cells demonstrated that INSR facilitates cell cycle transition from G1 to S phase and promotes AML cell proliferation. Furthermore, INSR upregulates vimentin and N-cadherin expression, thereby enhancing cell invasion and migration. By integrating transcriptome sequencing data with gene expression profile interactive analysis, we discovered that AKT1 expression levels were positively correlated with INSR expression, while AKT1 expression exhibited a negative correlation with the prognosis of AML patients. AKT1 expression inhibition reduced the proliferation and migratory activity of AML cells. Additionally, suppressing AKT1 expression diminished the impact of INSR on promoting AML cells proliferation, invasion, and migration. This study indicates that INSR expression is elevated in AML cells after treating with chidamide and that INSR promotes AML cells proliferation and migration by upregulating AKT1 expression.
{"title":"INSR/AKT1 axis promotes cells proliferation and migration in acute myeloid leukemia","authors":"Liman Lin , Chunle Zhao , Xiaoya Cai , Aiguo Liu , Min Xiao , Liting Chen , Dengju Li","doi":"10.1016/j.bbadis.2025.168132","DOIUrl":"10.1016/j.bbadis.2025.168132","url":null,"abstract":"<div><div>Chemotherapy resistance represents a major challenge in relapsed and refractory acute myeloid leukemia (AML). Therefore, it is necessary to investigate the mechanisms underlying chemotherapy resistance in AML. INSR not only highly expressed in AML cells treated with chidamide, but also elevated in AML patients. Subsequent overexpression and knockdown experiments of INSR in AML cells demonstrated that INSR facilitates cell cycle transition from G1 to S phase and promotes AML cell proliferation. Furthermore, INSR upregulates vimentin and N-cadherin expression, thereby enhancing cell invasion and migration. By integrating transcriptome sequencing data with gene expression profile interactive analysis, we discovered that AKT1 expression levels were positively correlated with INSR expression, while AKT1 expression exhibited a negative correlation with the prognosis of AML patients. AKT1 expression inhibition reduced the proliferation and migratory activity of AML cells. Additionally, suppressing AKT1 expression diminished the impact of INSR on promoting AML cells proliferation, invasion, and migration. This study indicates that INSR expression is elevated in AML cells after treating with chidamide and that INSR promotes AML cells proliferation and migration by upregulating AKT1 expression.</div></div>","PeriodicalId":8821,"journal":{"name":"Biochimica et biophysica acta. Molecular basis of disease","volume":"1872 3","pages":"Article 168132"},"PeriodicalIF":4.2,"publicationDate":"2025-12-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145764502","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}