1
|
Wu S, Ma X, Zhang X, Du K, Shi C, Almaamari AA, Han B, Su S, Liu Y. Knockdown of NDUFAF6 inhibits breast cancer progression via promoting mitophagy and apoptosis. Cancer Biol Ther 2025; 26:2445220. [PMID: 39706687 DOI: 10.1080/15384047.2024.2445220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 12/04/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND While NDUFAF6 is implicated in breast cancer, its specific role remains unclear. METHODS The expression levels and prognostic significance of NDUFAF6 in breast cancer were assessed using The Cancer Genome Atlas, Gene Expression Omnibus, Kaplan-Meier plotter and cBio-Portal databases. We knocked down NDUFAF6 in breast cancer cells using small interfering RNA and investigated its effects on cell proliferation and migration ability. We performed gene expression analysis and validated key findings using protein analysis. We also assessed mitochondrial activity and cellular metabolism. RESULTS NDUFAF6 was highly expressed in breast cancer, which was associated with a poorer prognosis. Knockdown of NDUFAF6 reduced the proliferation and migration ability of breast cancer cells. Transcriptome analysis revealed 2,101 differentially expressed genes enriched in apoptosis and mitochondrial signaling pathways. Western blot results showed NDUFAF6 knockdown enhanced apoptosis. In addition, differential gene enrichment analysis was related to mitochondrial signaling pathways, and western blot results verified that mitophagy was enhanced in NDUFAF6 knockdown breast cancer cells. JC-1 assay also showed that mitochondrial dysfunction and reactive oxygen species content were increased after knocking down NDUFAF6. In addition, basal and maximal mitochondrial oxygen consumption decreased, and intracellular glycogen content increased. CONCLUSIONS Knockdown of NDUFAF6 resulted in apoptosis and mitophagy in breast cancer cells and NDUFAF6 may be a potential molecular target for breast cancer therapy.
Collapse
Affiliation(s)
- Shang Wu
- Department of Breast Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Hebei Medical University, Shijiazhuang, China
| | - Xindi Ma
- Department of Breast Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Hebei Medical University, Shijiazhuang, China
| | - Xiangmei Zhang
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Hebei Medical University, Shijiazhuang, China
- Department of Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Kaiye Du
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Hebei Medical University, Shijiazhuang, China
- Radiotherapy Department, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Chao Shi
- Department of Breast Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Hebei Medical University, Shijiazhuang, China
| | - Ahmed Ali Almaamari
- The Key Laboratory of Neural and Vascular Biology, The Key Laboratory of New Drug Pharmacology and Toxicology, Department of Pharmacology, Ministry of Education, Hebei Medical University, Shijiazhuang, China
| | - Boye Han
- The Key Laboratory of Neural and Vascular Biology, The Key Laboratory of New Drug Pharmacology and Toxicology, Department of Pharmacology, Ministry of Education, Hebei Medical University, Shijiazhuang, China
| | - Suwen Su
- The Key Laboratory of Neural and Vascular Biology, The Key Laboratory of New Drug Pharmacology and Toxicology, Department of Pharmacology, Ministry of Education, Hebei Medical University, Shijiazhuang, China
| | - Yunjiang Liu
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Hebei Medical University, Shijiazhuang, China
- The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
2
|
Oğlak SC, Aşır F, Yılmaz EZ, Aşır A, Bolluk G, Korak T, Ayaz H, Ağaçayak E. Immunohistochemical and bioinformatics analysis of the placental BCL-2 and Beclin-1 expressions in preeclampsia and HELLP syndrome. J OBSTET GYNAECOL 2025; 45:2478577. [PMID: 40094536 DOI: 10.1080/01443615.2025.2478577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 03/07/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND This study aimed to examine the alterations in apoptosis and autophagy in placental tissues from normal pregnancies compared to those affected by preeclampsia (PE) and HELLP (haemolysis, elevated liver enzymes, low platelets) syndrome. We analysed the expression of autophagy-associated proteins, Beclin-1 and BCL-2, in human placental tissues and assessed their variations in placentas from pregnancies complicated by PE and HELLP syndrome by immunohistochemical (IHC) and in silico analyses. METHODS An experimental case-control study was performed, involving 40 pregnant women complicated with preeclampsia, 25 pregnant women with HELLP syndrome, and 40 healthy pregnant women. The placental sections were stained with BCL-2 and Beclin-1 immunostains and subjected to IHC examination. The results of the IHC staining were assessed using semi-quantitative analysis. In silico analyses were performed using STRING and Cytoscape software to construct protein interaction networks for BCL-2 and Beclin-1 in PE and HELLP syndrome, followed by Gene Ontology analysis of common interactors to identify significant biological pathways. RESULTS Both BCL-2 expression was significantly decreased (p < 0.001 and p < 0.0001, respectively) and Beclin-1 staining was significantly increased (p < 0.0001 and p < 0.0001, respectively) in the PE group and HELLP group compared to the control group. The changes in BCL-2 and Beclin-1 expression between PE and the HELLP group were also statistically significant. BCL2 expression was notably lower (p < 0.0001), and Beclin-1 staining was significantly higher (p < 0.05) in the HELLP group compared to the PE group. In PE, BCL-2 interactors were enriched in apoptosis, cytokine production, and cell proliferation pathways, while Beclin-1 interactors were linked to autophagy and phosphatidylinositol-mediated signalling. In HELLP, BCL-2 interactors were involved in inflammatory response regulation, whereas Beclin-1 interactors were associated with vascular endothelial growth factor (VEGF) signalling and immune regulation. CONCLUSIONS The differential expression patterns of BCL-2 and Beclin-1 between the PE and HELLP groups suggest that these proteins play distinct roles in the pathophysiology of these conditions.
Collapse
Affiliation(s)
- Süleyman Cemil Oğlak
- Department of Obstetrics and Gynaecology, Health Sciences University, Gazi Yaşargil Training and Research Hospital, Diyarbakır, Turkey
| | - Fırat Aşır
- Department of Histology and Embryology, Dicle University Faculty of Medicine, Diyarbakır, Turkey
| | - Emine Zeynep Yılmaz
- Department of Obstetrics and Gynaecology, Bahçelievler Memorial Hospital, Istanbul, Turkey
| | - Ayşegül Aşır
- Department of Pediatrics, Health Sciences University, Gazi Yaşargil Training and Research Hospital, Diyarbakır, Turkey
| | - Gökhan Bolluk
- Department of Perinatology, Başakşehir Çam and Sakura City Hospital, Istanbul, Turkey
| | - Tuğcan Korak
- Department of Medical Biology, Kocaeli University Faculty of Medicine, Kocaeli, Turkey
| | - Hayat Ayaz
- Department of Histology and Embryology, Dicle University Faculty of Medicine, Diyarbakır, Turkey
| | - Elif Ağaçayak
- Department of Obstetrics and Gynaecology, Dicle University Faculty of Medicine, Diyarbakır, Turkey
| |
Collapse
|
3
|
Jia LL, Wu CJ, Ye PW, Zhang Q, Liu H, Li TP, Hu XL. Terrestrosin D promotes autophagy and apoptosis of breast cancer cells through PSMD1-dependent activation of P53 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156883. [PMID: 40412055 DOI: 10.1016/j.phymed.2025.156883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/30/2025] [Accepted: 05/19/2025] [Indexed: 05/27/2025]
Abstract
BACKGROUND AND PURPOSE Breast cancer, particularly triple-negative breast cancer (TNBC), poses a significant threat to women's health. In tumor cells, autophagy and apoptosis are double-edged swords, playing complex roles in cancer progression and treatment. This study aimed to investigate whether Terrestrosin D (TED) exerts antitumor effects on TNBC by modulating autophagy and apoptosis, and to elucidate the underlying molecular mechanisms. METHODS The antiproliferative and pro-apoptotic effects of TED on TNBC cells were assessed using CCK-8, EdU assay, Live/Dead staining, and flow cytometry. Autophagy was monitored through immunofluorescence and confocal microscopy. RNA sequencing was performed to identify the pathways and molecular targets involved. The anti-TNBC effects of TED were further evaluated in vivo using tumor xenograft models. Western blotting was conducted to validate the relationship between PSMD1, P53, and TED-induced antitumor activity. RESULTS TED exhibited significant antitumor effects both in vitro and in vivo. Cellular phenotypic analyses revealed that TED promoted autophagy and apoptosis. Transcriptomic analyses indicated that TED stabilizes P53 expression and activates the P53 signaling pathway by inhibiting the function of PSMD1. CONCLUSION TED exhibits potent antitumor effects on TNBC by promoting autophagy and apoptosis. It achieves this through PSMD1 inhibition, stabilizing P53 expression, and activating the P53 pathway. Notably, this study is the first to demonstrate that TED directly targets PSMD1, a key proteasomal regulator, thereby unveiling a novel mechanism for P53 stabilization in TNBC. These findings provide new insights into the therapeutic modulation of the PSMD1 - P53 axis by natural compounds and support the development of TED as a multi-functional agent for aggressive breast cancers.
Collapse
Affiliation(s)
- Li-Ling Jia
- Department of Anaesthesia, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, PR China
| | - Cheng-Jie Wu
- Breast Center, Department of General Surgery, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, 510515, PR China
| | - Pei-Wen Ye
- Breast Center, Department of General Surgery, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, 510515, PR China
| | - Qian Zhang
- Breast Center, Department of General Surgery, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, 510515, PR China
| | - Hua Liu
- Department of Anaesthesia, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, PR China
| | - Tu-Ping Li
- Department of Anaesthesia, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, PR China
| | - Xiao-Lei Hu
- Breast Center, Department of General Surgery, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, 510515, PR China.
| |
Collapse
|
4
|
Chao P, Zhang X, Zhang L, Han Z, Jie R, Duan P, Cao M, Yang A. Electroacupuncture as a promising therapeutic strategy for doxorubicin-induced heart failure: Insights into the PI3K/AKT/mTOR/ULK1 and AMPK /mTOR /ULK1 pathways. Colloids Surf B Biointerfaces 2025; 251:114590. [PMID: 40024111 DOI: 10.1016/j.colsurfb.2025.114590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 02/19/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND Electroacupuncture (EA), a traditional Chinese medicine therapy, exhibits cardioprotective and therapeutic effects against cardiac injury. However, the precise mechanisms underlying these benefits remain unclear. PURPOSE The aim of this study is to examine the impact of EA on Doxorubicin-Induced heart failure and elucidate the mechanisms involved. METHODS C57BL/6 mice were randomly assigned to six experimental groups, including a control group, a DCM group, a DCM group receiving non-acupoint EA (NEA), and a DCM group receiving acupoint EA (EA). The cardiac function, levels of inflammatory factors, and markers of apoptosis were assessed both in vivo and in vitro. The presence of AMPK/mTOR/ULK1(Ser317) and PI3K/AKT/mTOR/ULK1(Ser757) was confirmed. RESULTS EA stimulation significantly improved cardiac function, as evidenced by increased left ventricular ejection fraction (LVEF), E/A ratio, and fractional shortening (FS%) compared to the DCM group (p < 0.05). After EA stimulation, the phosphorylation levels of PI3K/AKT increase, leading to elevated expression of mTOR/ULK1(Ser757), which ultimately inhibited the expression of apoptosis-related proteins and inflammatory factors. Simultaneously, EA stimulation could inhibit the phosphorylation levels of AMPK, reducing the expression of mTOR/ULK1(Ser317), and thereby also inhibiting the expression of apoptosis-related proteins and inflammatory factors. CONCLUSIONS This study showed that EA stimulation can counteract myocardial damage caused by apoptosis and inflammation, thereby significantly improving cardiac function and prognosis in HF mice. The mechanism may be that EA stimulation activates the PI3K/AKT/mTOR/ULK1(ser757) pathway and inhibits the AMPK/ULK1(ser317) pathway. EA stimulation exerts the same effect by regulating these two pathways in different directions, ultimately reducing myocardial cell apoptosis and cardiac inflammation.
Collapse
Affiliation(s)
- Peng Chao
- People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Xueqin Zhang
- People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Lei Zhang
- People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Zhengyang Han
- People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Runda Jie
- People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Pingxiu Duan
- People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Min Cao
- People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Aiping Yang
- People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, China.
| |
Collapse
|
5
|
Guo Z, He L, Wang W, Tian S, Lin R. FUT2-dependent fucosylation of LAMP1 promotes the apoptosis of colorectal cancer cells by regulating the autophagy-lysosomal pathway. Cancer Lett 2025; 619:217643. [PMID: 40112906 DOI: 10.1016/j.canlet.2025.217643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/06/2025] [Accepted: 03/15/2025] [Indexed: 03/22/2025]
Abstract
Fucosyltransferase 2 (FUT2) is an enzyme that adds fucose to proteins or lipids via α-1,2-fucosylation in the intestinal mucosa. While FUT2 deficiency is linked to increased susceptibility to inflammatory bowel disease (IBD), its role in colorectal cancer (CRC) is unclear, and the molecular mechanisms involved remain largely unknown. We established an azoxymethane (AOM) and dextran sulfate sodium (DSS) model to induce CRC. FUT2 expression was assessed in human CRC tissues, AOM/DSS-induced mouse models, and CRC cell lines using qRT-PCR, western blotting, and UEA-I staining. FUT2 knockout (FUT2△IEC) mice were treated with AOM/DSS, and FUT2-overexpressing CRC cells were created to evaluate the effects of FUT2 on apoptosis in both in vitro and in vivo settings through Western blot analyses and functional assays. N-glycoproteomics, UEA-I chromatography, and co-immunoprecipitation were utilized to identify regulatory mechanisms and target fucosylated proteins. FUT2 expression and α-1,2-fucosylation were significantly decreased in CRC. FUT2 deficiency worsened AOM/DSS-induced CRC and reduced tumor apoptosis, while FUT2 overexpression induced apoptosis and inhibited proliferation in CRC cells and xenografts. Mechanistically, FUT2 appears to suppress autophagy by impairing lysosomal function and directly targeting and fucosylating LAMP1, contributing to lysosomal dysfunction. Our study reveals a fucosylation-dependent antitumor mechanism of FUT2 in CRC, suggesting potential therapeutic strategies for CRC treatment.
Collapse
Affiliation(s)
- Zijun Guo
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lingnan He
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong New Area, Shanghai, China
| | - Weijun Wang
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuxin Tian
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rong Lin
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
6
|
Qusty NF, Bokhari BT, Taha M, Alobaidy MA, Al-Kushi AG, Sembawa HA, Abdelbagi O, Baokbah TAS, Obaid R, Albar HT, Babateen O, Dahran N. Empagliflozin Inhibits Cadmium-Induced Hepatic Cell Apoptosis Through Endoplasmic Reticulum Stress and Autophagy Pathways. Biol Trace Elem Res 2025:10.1007/s12011-025-04631-z. [PMID: 40372601 DOI: 10.1007/s12011-025-04631-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 04/16/2025] [Indexed: 05/16/2025]
Abstract
Cadmium (Cd), a well-known toxic heavy metal, adversely affects multiple organs. The SGLT-2 inhibitor empagliflozin (EMPA) exhibits significant antioxidant properties and hypoglycemic potential. This study aimed to investigate the hepatoprotective effect of EMPA against Cd-induced liver injury and elucidate its molecular mechanisms. Thirty-two male rats were allocated into four groups of eight rats each: group I (control group), group II (EMPA group), group III (Cd group), and group IV (Cd + EMPA group). Cd intake disrupted liver enzymes (ALT, AST, and ALP) and impaired hepatic histological architecture. Cd induced hepatic oxidative stress, as evidenced by increased MDA levels and reduced antioxidant enzymes, including SOD, GPx, and CAT. It downregulated the Nrf2/HO-1 pathway and elevated proinflammatory mediators IL-1β, IL-6, and TNF-α. Furthermore, Cd increased ER stress markers GRP78 and CHOP, along with apoptotic markers Bax and caspase-3 while decreasing anti-apoptotic Bcl-2 and reducing the autophagic indicator Beclin-1. Interestingly, EMPA administration in the Cd + EMPA group attenuated Cd-induced hepatic deterioration, improving hepatocyte structure. This beneficial effect was driven by the downregulation of hepatic oxidative stress, inflammation, ER stress, and apoptosis, alongside the upregulation of the autophagy process. In conclusion, this study highlights the hepatoprotective effect of EMPA against Cd-induced liver injury, elucidating its underlying molecular mechanisms.
Collapse
Affiliation(s)
- Naeem F Qusty
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al‒Qura University, Makkah, 21955, Saudi Arabia
| | - Bayan T Bokhari
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al‒Qura University, Makkah, 21955, Saudi Arabia
| | - Medhat Taha
- Department of Anatomy, Al-Qunfudah Medical College, Umm Al-Qura University, Al-Qunfudhah, Saudi Arabia.
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| | - Mohammad Ahmad Alobaidy
- Department of Anatomy, Faculty of Medicine, Umm Al-Qura University, Makkah, P.O. Box 7607, Saudi Arabia
| | - Abdullah G Al-Kushi
- Department of Anatomy, Faculty of Medicine, Umm Al-Qura University, Makkah, P.O. Box 7607, Saudi Arabia
| | - Hatem A Sembawa
- Department of Surgery, Faculty of Medicine, Umm Al-Qura University, Holy Makkah, Saudi Arabia
| | - Omer Abdelbagi
- Department of Pathology, Qunfudah Faculty of Medicine, Umm-Al-Qura University, Makkah, Kingdom of Saudi Arabia
| | - Tourki A S Baokbah
- Department of Medical Emergency Services, College of Health Sciences-AlQunfudah, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Rami Obaid
- Department of Medical Genetics, Faculty of Medicine, -Qunfudah, Umm Al-Qura University, Al-Qunfudhah, Saudi Arabia
| | - Halah Tariq Albar
- Department of Physiology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Omar Babateen
- Department of Physiology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Naief Dahran
- Department of Basic Medical Sciences, College of Medicine, University of Jeddah, Jeddah, Saudi Arabia
| |
Collapse
|
7
|
Bao C, Zhang Y, Feng J, Hong X, Gao N, Feng G. Deciphering tuberculosis: lysosome-centric insights into pathogenesis and therapies. Front Cell Infect Microbiol 2025; 15:1582037. [PMID: 40438237 PMCID: PMC12116394 DOI: 10.3389/fcimb.2025.1582037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Accepted: 04/17/2025] [Indexed: 06/01/2025] Open
Abstract
Tuberculosis is a widely spread disease caused by Mycobacterium tuberculosis (Mtb). The pathogenicity of the pathogen is closely associated with the immune defense mechanisms of the host cells. As key cellular degradation and metabolic centers, lysosomes critically regulate tuberculosis infection. When Mtb invades the host, it is taken up by macrophages and enters phagosomes. Subsequently, the phagosomes fuse with lysosomes and form phagolysosomes, which eliminate the pathogenic bacteria through the acidic environment and hydrolytic enzymes within lysosomes. However, Mtb can interfere with the normal functions of lysosomes through various strategies. It can secrete specific factors (such as ESAT-6, ppk-1, and AcpM) to inhibit the acidification of lysosomes, enzyme activity, and the fusion of phagosomes and lysosomes, thereby enabling Mtb proliferation within host cells. An in-depth exploration of the mechanism of the interaction between Mtb and lysosomes will both uncover bacterial immune evasion strategies and identify novel anti-tuberculosis therapeutic targets.
Collapse
Affiliation(s)
- Cui Bao
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Respiratory and Critical Care Medicine, The Second Clinical Medical School of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuanyuan Zhang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Respiratory and Critical Care Medicine, The Second Clinical Medical School of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiao Feng
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Respiratory and Critical Care Medicine, The Second Clinical Medical School of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiuwen Hong
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Respiratory and Critical Care Medicine, The Second Clinical Medical School of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Nan Gao
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Respiratory and Critical Care Medicine, The Second Clinical Medical School of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ganzhu Feng
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
8
|
Xie S, Wang Y, Zhao C, Cui Y, Gu C, Wu W. Putrescine promotes maturation of oocytes from reproductively old mice via mitochondrial autophagy. Reprod Biomed Online 2025; 50:104495. [PMID: 40068351 DOI: 10.1016/j.rbmo.2024.104495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 09/25/2024] [Accepted: 10/14/2024] [Indexed: 05/12/2025]
Abstract
RESEARCH QUESTION Does putrescine (PUT) improve oocytes from reproductively old mice by promoting mitochondrial autophagy? DESIGN Germinal vesicle stage cumulus-oocyte complexes (COCs) were obtained from 9-month old female C57BL/6N mice and divided into control, PUT and difluoromethylornithine, inhibitor (DFMO) groups. These germinal vesicle COCs underwent mouse in-vitro maturation (IVM) culture to observe the extrusion of the first polar body in each group. Using JC-1, dichloro-dihydro-fluorescein diacetate fluorescent probes and a confocal microscope, the mitochondrial membrane potential integrity and reactive oxygen species levels were measured in metaphase II stage oocytes. The expression and cellular localization of the p53 protein were examined by immunofluorescence. Reverse transcription quantitative polymerase chain reaction was used to detect the activation of mitochondrial autophagy pathways. The potential mechanisms through which PUT improves oocytes from reproductively old mice were explored by single-cell transcriptomic analysis. Autophagosomes, autolysosomes and mitochondria in different groups were directly observed using transmission electron microscopy. RESULTS The addition of exogenous PUT can promote IVM of oocytes from reproductively old mice. It reduces oxidative stress by promoting the autophagy of damaged mitochondria, decreasing the levels of reactive oxygen species and increasing mitochondrial membrane potential. It affects the expression and subcellular localization of the p53 protein, and increases the expression of transcription factor EB, which may be the potential mechanism behind its promotion of autophagy. CONCLUSION The target and regulatory pathway of PUT in oocytes was clarified. Putrescine is an effective small molecule compound with significant potential for non-invasively improving the fertility of elderly women.
Collapse
Affiliation(s)
- Siyuan Xie
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China.; School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yuyi Wang
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Chenxi Zhao
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Yugui Cui
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Chunyan Gu
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Wei Wu
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China..
| |
Collapse
|
9
|
Wen Y, Zou Z, Li Y, Zhang D, Liu Z, Liu H, Li X, Wu W, Zeng L, Zou Q, Yi W. Dendrimer nanocarriers for targeted co-delivery of MiR-146b-3p and 5-ALA to synergistic photodynamic therapy in secondary hyperparathyroidism. Int J Biol Macromol 2025; 310:143307. [PMID: 40253027 DOI: 10.1016/j.ijbiomac.2025.143307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 04/21/2025]
Abstract
The combination of photodynamic therapy (PDT) and gene therapy is a viable therapeutic approach for the management of secondary hyperparathyroidism (SHPT). Nevertheless, the effective delivery of photosensitizers or nucleotide drugs remains a crucial limitation in achieving therapeutic efficacy. MiR-146b-3p directly targeted BCL2, and its overexpression enhanced the antiproliferative, proautophagic, and proapoptotic effects during 5-aminolevulinic acid (5-ALA)-mediated PDT in this study. Herein, we investigated the potential of codelivering 5-ALA and miR-146b-3p to SHPT primary cells via polyamidoamine (PAMAM) and achieving enhanced therapeutic efficacy relative to that of monotreatment. The fabrication of the PAMAM-based 5-ALA and miR-146b-3p dual-delivery system (5-ALA@PAMAM/miR) involved the use of covalent condensation reactions and electrostatic self-assembly. The nanoparticles were characterized by various analytical techniques, including transmission electron microscopy (TEM), zeta potential measurements, and size measurements. Fluorescence and confocal laser scanning microscopy demonstrated a greater degree of cellular uptake of nanoparticles. Moreover, the synthesized nanoparticles considerably enhanced the effectiveness of PDT without systemic toxicity both in vitro and in vivo. Overall, the nanocarrier-gene-photosensitizer coloaded system is a promising platform for the efficient simultaneous delivery of miR-146b-3p and 5-ALA and achieves synergistic therapeutic effects.
Collapse
Affiliation(s)
- Ying Wen
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Clinical Research Center For Breast Disease In Hunan Province, Changsha, Hunan 410011, China
| | - Zhen Zou
- Laboratory of Chemical Biology&Traditional Chinese Medicine Research Ministry of Education, College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, Hunan, 410114, China
| | - Yitong Li
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Clinical Research Center For Breast Disease In Hunan Province, Changsha, Hunan 410011, China
| | - Danhua Zhang
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Clinical Research Center For Breast Disease In Hunan Province, Changsha, Hunan 410011, China
| | - Ziru Liu
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Clinical Research Center For Breast Disease In Hunan Province, Changsha, Hunan 410011, China
| | - Hong Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Xiejia Li
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Wei Wu
- Department of General Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Liyun Zeng
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Clinical Research Center For Breast Disease In Hunan Province, Changsha, Hunan 410011, China.
| | - Qiongyan Zou
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Clinical Research Center For Breast Disease In Hunan Province, Changsha, Hunan 410011, China.
| | - Wenjun Yi
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Clinical Research Center For Breast Disease In Hunan Province, Changsha, Hunan 410011, China.
| |
Collapse
|
10
|
Wang R, Yan J, Zhang H, Zhu X, Xie D, Wang T, Li X. New insights into heavy metal cadmium-induced liver injury: Prominent role of programmed cell death mechanisms. Toxicology 2025; 517:154169. [PMID: 40318836 DOI: 10.1016/j.tox.2025.154169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/30/2025] [Accepted: 04/30/2025] [Indexed: 05/07/2025]
Abstract
The heavy metal cadmium (Cd) is an important environmental factor that induces liver injury and contributes to liver disease. Ongoing research aims to refine our understanding of the pathogenesis of cadmium-induced liver injury and the interactions between the various mechanisms. Oxidative stress, described as a pathophysiological basis of liver injury, is a process in which reactive oxygen species are generated, causing the destruction of hepatocyte structure and cellular dysfunction. Additionally, the activation of oxidative stress downstream signals regulates several forms of cell death, such as apoptosis, necroptosis, autophagy, ferroptosis, and pyroptosis, which significantly contributes to liver damage. Furthermore, the interplay between different types of programmed cell death highlights the complexity of liver injury mechanisms. This review summarizes the role of programmed cell death in Cd-induced liver injury and explores the relationships between different programmed cell death pathways, which is expected to provide new insights into the mechanisms of Cd-induced liver injury.
Collapse
Affiliation(s)
- Ruipeng Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China
| | - Jun Yan
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China; Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China; Key Laboratory Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou 730000, China; Cancer Prevention and Treatment Center of Lanzhou University School of Medicine, Lanzhou 730000, China; Hepatopancreatobiliary Surgery Institute of Gansu Province, Lanzhou 730000, China
| | - Honglong Zhang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China
| | - Xingwang Zhu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China
| | - Danna Xie
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China
| | - Tingting Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China
| | - Xun Li
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China; Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China; Key Laboratory Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou 730000, China; Cancer Prevention and Treatment Center of Lanzhou University School of Medicine, Lanzhou 730000, China; Hepatopancreatobiliary Surgery Institute of Gansu Province, Lanzhou 730000, China; Clinical Research Center for General Surgery of Gansu Province, Lanzhou 730000, China.
| |
Collapse
|
11
|
Zhao G, Qi J, Li F, Ma H, Wang R, Yu X, Wang Y, Qin S, Wu J, Huang C, Ren H, Zhang B. TRAF3IP3 Induces ER Stress-Mediated Apoptosis with Protective Autophagy to Inhibit Lung Adenocarcinoma Proliferation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411020. [PMID: 40068093 PMCID: PMC12061266 DOI: 10.1002/advs.202411020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 02/17/2025] [Indexed: 05/10/2025]
Abstract
TNF receptor-associated factor 3 interacting protein 3 (TRAF3IP3/T3JAM) exhibits dual roles in cancer progression. While upregulated in most malignancies and critical for immune regulation. However, the specific effects and molecular mechanisms of TRAF3IP3 on the progression of lung adenocarcinoma (LUAD) remains poorly understood. This study reveals TRAF3IP3 is upregulated in several tumor tissues but exclusively decreased in LUAD and Lung squamous cell carcinoma (LUSC) tissues, consequential in a favorable overall survival (OS) in LUAD rather than LUSC. Herein, it is reported that TRAF3IP3 can suppress cell proliferation and promote the apoptosis rate of LUAD cells by inducing excessive ER stress-related apoptosis. Importantly, TRAF3IP3 triggers ER stress via the PERK/ATF4/CHOP pathway, accompanied by stimulated ER stress-induced cytoprotective autophagy in LUAD cells. Through IP-MS analysis, STRN3 is identified as a direct downstream interactor with TRAF3IP3 and corroborated to regulate ER stress positively. Mechanistically, TRAF3IP3 facilitates the recruitment of STRN3 to the ER lumen through its transmembrane domain and fulfills its functional role in ER stress in an STRN3-dependent manner in LUAD cells. Given its dual role in orchestrating ER stress-associated apoptosis and autophagy in LUAD cell fate determination, the importance of TRAF3IP3 is highlighted as novel therapeutic target for LUAD treatment.
Collapse
Affiliation(s)
- Guang Zhao
- Department of Thoracic Surgerythe First Affiliated Hospital of Xi'an Jiaotong University277 West Yanta Road, Xi'anXi'anShaanxi710061China
- Department of Thoracic SurgerySichuan Provincial People's Hospital: Sichuan Academy of Medical Sciences and Sichuan People's HospitalChengduSichuan610072China
| | - Jun Qi
- Department of DermatologyGansu Provincial Maternity and Child‐care Hospital (Gansu Provincial Central Hospital)Lan ZhouGansu730079China
| | - Fang Li
- Institute of Basic Medical SciencesXi'an Medical UniversityNo.1 XinWang Road, Weiyang DistrictXi'anShaanxi710021China
| | - Haotian Ma
- Health Science CenterXi'an Jiaotong UniversityXi'an710061China
| | - Rui Wang
- Department of Thoracic Surgerythe First Affiliated Hospital of Xi'an Jiaotong University277 West Yanta Road, Xi'anXi'anShaanxi710061China
| | - Xiuyi Yu
- Department of Thoracic Surgerythe First Affiliated Hospital of Xiamen UniversityXiamen361003China
| | - Yufei Wang
- Health Science CenterXi'an Jiaotong UniversityXi'an710061China
| | - Sida Qin
- Department of Thoracic Surgerythe First Affiliated Hospital of Xi'an Jiaotong University277 West Yanta Road, Xi'anXi'anShaanxi710061China
| | - Jie Wu
- Department of Radiation OncologyShaanxi Provincial People's HospitalXi'anShaanxi710061China
| | - Chen Huang
- Department of Cell Biology and GeneticsSchool of Basic Medical SciencesXi'an Jiaotong University Health Science CenterXi'anShaanxi710061China
| | - Hong Ren
- Department of Thoracic Surgerythe First Affiliated Hospital of Xi'an Jiaotong University277 West Yanta Road, Xi'anXi'anShaanxi710061China
| | - Boxiang Zhang
- Department of Thoracic Surgerythe First Affiliated Hospital of Xi'an Jiaotong University277 West Yanta Road, Xi'anXi'anShaanxi710061China
| |
Collapse
|
12
|
Lu Y, Liu Y, Cao J, Zhang Y, Zheng Y, Wang F. Waterborne ammonia toxicity damages crustacean hemocytes via lysosome-dependent autophagy: A case study of swimming crabs Portunus trituberculatus. ENVIRONMENTAL RESEARCH 2025; 272:120985. [PMID: 39983961 DOI: 10.1016/j.envres.2025.120985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 02/23/2025]
Abstract
Waterborne ammonia is a threat to animal health and its accumulation is typical of aquatic ecosystems. Autophagy serves as a safeguard of intracellular homeostasis, yet its role in maintaining the health of hemocytes, the master regulators of crustacean immunity, remains unclear. Herein, the swimming crab (Portunus trituberculatus) is employed as a case study to illustrate the impact of ammonia on hemocyte health via autophagy. This study showed the occurrence of abnormal cellular structure and significant accumulation of malondialdehyde (MDA) and reactive oxygen species (ROS) (P < 0.05), demonstrating that severe ammonia stress can damage hemocytes. This was accompanied by significant increase of autophagy hemocytes fraction and apoptosis (P < 0.05). Meanwhile, there was a significant increase in the expression of Beclin1 and microtubule-associated protein 1 light chain 3 (LC3-II) (P < 0.05). This suggests an ammonia-induced autophagy initiation. However, ammonia stress significantly decreased lysosomal fluorescence intensity (P < 0.05) and expression of the marker gene lysosomal-associated membrane protein 1 (LAMP1) (P < 0.05). These imply an ammonia-induced repression of lysosome-dependent autophagy degradation, which may underlie the pronounced increase in apoptosis (P < 0.05). After the administration of the autophagy activator rapamycin (Rap), rather than the inhibitor 3-Methyladenine (3-MA), the levels of apoptosis, ROS and the fraction of autophagic cells were significantly decreased (P < 0.05), demonstrating a mitigation of the ammonia-induced cell damage through lysosome-dependent autophagy degradation. This study sheds light on how crustaceans respond to ammonia exposure by demonstrating the significance of lysosome-dependent autophagy in maintaining hemocyte health.
Collapse
Affiliation(s)
- Yunliang Lu
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Yingying Liu
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Jianwei Cao
- Key Laboratory of Mariculture of Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Yueqi Zhang
- Key Laboratory of Mariculture of Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Yuan Zheng
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China.
| | - Fang Wang
- Key Laboratory of Mariculture of Ministry of Education, Ocean University of China, Qingdao, 266003, China.
| |
Collapse
|
13
|
Li L, Gu L, Hua Y, Xu H, Ling Y, Tong L, Chen M, Ma R. Kumatakenin alleviates depressive-like behaviors by suppressing excessive autophagy in hippocampus via ATG5. Eur J Pharmacol 2025; 999:177688. [PMID: 40294778 DOI: 10.1016/j.ejphar.2025.177688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 04/25/2025] [Accepted: 04/25/2025] [Indexed: 04/30/2025]
Abstract
The prevalence of depression has been escalating annually, imposing a substantial burden on both individuals and society. Radix Astragali, a multifaceted herb utilized in traditional Chinese medicine, has been employed in clinical settings for the treatment of depression. Kumatakenin, a principal active constituent of Radix Astragali, has yet to be thoroughly investigated for its potential antidepressant effects. In this study, we elucidated a novel effect of Kumatakenin, which ameliorated depression-like behaviors by modulating excessive autophagy in the hippocampus of mice exhibiting depressive symptoms. Utilizing transmission electron microscopy, we identified a significant increase in autophagosomes within the hippocampal region of depressed mice, which was notably reduced following Kumatakenin administration. Further, molecular docking analyses revealed an interaction between Kumatakenin and autophagy-related gene 5 (ATG5). At the protein level, Kumatakenin administration resulted in a marked decrease in microtubule-associated protein 1 light chain 3 (LC3) and ATG5 levels within hippocampal tissues. At the cellular level, in a corticosterone (CORT)-induced cell model, Kumatakenin significantly diminished the levels of LC3 and ATG5. Upon overexpression of ATG5 through lentiviral transfection, the ability of Kumatakenin to reduce LC3 and ATG5 levels was nullified. These observations suggested that Kumatakenin exerted its antidepressant effects by decreasing LC3 and ATG5 concentrations in hippocampal tissues.
Collapse
Affiliation(s)
- Lei Li
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| | - Ling Gu
- School of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China.
| | - Yang Hua
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| | - Huiying Xu
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| | - Yuyan Ling
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| | - Lijun Tong
- Medical Laboratory Department, Inner Mongolia Autonomous Region Mental Health Center, Hohhot, 010010, PR China.
| | - Meijuan Chen
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| | - Ruiting Ma
- Medical Laboratory Department, Inner Mongolia Autonomous Region Mental Health Center, Hohhot, 010010, PR China.
| |
Collapse
|
14
|
朱 生, 李 忠. [Mechanism of extracellular vesicles in the repair of intervertebral disc degeneration]. SHENG WU YI XUE GONG CHENG XUE ZA ZHI = JOURNAL OF BIOMEDICAL ENGINEERING = SHENGWU YIXUE GONGCHENGXUE ZAZHI 2025; 42:409-416. [PMID: 40288986 PMCID: PMC12035626 DOI: 10.7507/1001-5515.202403046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 01/26/2025] [Indexed: 04/29/2025]
Abstract
Extracellular vesicles (EVs), defined as cell-secreted nanoscale vesicles that carry bioactive molecules, have emerged as a promising therapeutic strategy in tumor and tissue regeneration. Their potential in repairing intervertebral disc degeneration (IDD) through multidimensional regulatory mechanisms is a rapidly advancing field of research. This paper provided an overview of the mechanisms of EVs in IDD repair, thoroughly reviewed recent literature on EVs for IDD, domestically and internationally, and summarized their therapeutic mechanisms. In IDD repair, EVs could act through different mechanisms at the molecular, cellular, and tissue levels. At the molecular level, EVs could treat IDD by inhibiting inflammatory reactions, suppressing oxidative stress, and regulating the synthesis and decomposition of extracellular matrix. At the cellular level, EVs could treat IDD by inhibiting cellular pyroptosis, ferroptosis, and apoptosis and promoting cell proliferation and differentiation. At the tissue level, EVs could treat IDD by inhibiting neovascularization. EVs have a strong potential for clinical application in the treatment of IDD and deserve more profound study.
Collapse
Affiliation(s)
- 生旭 朱
- 大连医科大学附属第一医院 骨科(辽宁大连 116011)Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P. R. China
| | - 忠海 李
- 大连医科大学附属第一医院 骨科(辽宁大连 116011)Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P. R. China
| |
Collapse
|
15
|
Walweel N, Cinar V, Mersin O, Macit S, Yildiz U, Demirel E, Tunç CU, Ulutabanca H, Hamurcu Z, Yuksel Durmaz Y, Aydin O. Enhanced In Vitro and In Vivo Autophagy Suppression via LC3 siRNA-Loaded "Smart" Nanoparticles and Doxorubicin Combination Therapy in Triple Negative Breast Cancer. ACS APPLIED BIO MATERIALS 2025; 8:2938-2953. [PMID: 40056448 DOI: 10.1021/acsabm.4c01778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2025]
Abstract
Autophagy plays a complex role in cancer progression, serving as both a tumor suppressor and a promoter, depending on the context. In triple-negative breast cancer (TNBC), a particularly aggressive subtype with limited therapeutic options, autophagy inhibition has emerged as a promising strategy to enhance the efficacy of chemotherapy. This study investigates the synergistic effects of autophagy suppression using LC3 siRNA-loaded "smart" nanoparticles (LC3siRNA-NPs) in combination with doxorubicin (DOX) to overcome chemoresistance in TNBC. We engineered a well-defined copolymer, poly[hexyl methacrylate-co-2-(dimethylamino) ethyl methacrylate-co-trimethylaminoethyl methacrylate iodide], and a PEG heteroarm beta-cyclodextrin (βCD) core star copolymer that delivers LC3 siRNA, effectively silencing the autophagy-related gene LC3. In vitro, the coadministration of LC3siRNA-NPs and DOX significantly inhibited TNBC cell proliferation, migration, and colony formation, while inducing apoptosis more effectively than either treatment alone. Mechanistically, this combination downregulated key oncogenic markers such as PARP, cyclin D1, and Src, enhancing the therapeutic outcome. In vivo, treatment with LC3siRNA-NPs and DOX in a TNBC xenograft model resulted in superior tumor growth suppression compared to that with monotherapy alone. Our findings highlight the potential of autophagy-targeting nanocarriers to improve chemotherapy outcomes and provide an effective approach to TNBC treatment by enhancing chemotherapeutic sensitivity and reducing tumor resistance.
Collapse
MESH Headings
- Doxorubicin/pharmacology
- Doxorubicin/chemistry
- Triple Negative Breast Neoplasms/drug therapy
- Triple Negative Breast Neoplasms/pathology
- Triple Negative Breast Neoplasms/metabolism
- Humans
- Autophagy/drug effects
- Nanoparticles/chemistry
- RNA, Small Interfering/pharmacology
- RNA, Small Interfering/genetics
- RNA, Small Interfering/chemistry
- Animals
- Mice
- Female
- Cell Proliferation/drug effects
- Microtubule-Associated Proteins/genetics
- Microtubule-Associated Proteins/metabolism
- Microtubule-Associated Proteins/antagonists & inhibitors
- Biocompatible Materials/chemistry
- Biocompatible Materials/pharmacology
- Biocompatible Materials/chemical synthesis
- Drug Screening Assays, Antitumor
- Materials Testing
- Particle Size
- Apoptosis/drug effects
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/chemistry
- Mice, Nude
- Cell Line, Tumor
- Mice, Inbred BALB C
- Antibiotics, Antineoplastic/pharmacology
- Antibiotics, Antineoplastic/chemistry
- Dose-Response Relationship, Drug
Collapse
Affiliation(s)
- Nada Walweel
- Department of Biomedical Engineering, Erciyes University, Kayseri 38039, Turkey
- NanoThera Lab, ERFARMA-Drug Application and Research Center, Erciyes University, Kayseri 38280, Turkey
| | - Venhar Cinar
- Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri 38030, Turkey
- GENKOK-Betül-Ziya Eren Genome and Stem Cell Center, Erciyes University, Kayseri 38280, Turkey
| | - Osman Mersin
- Department of Biomedical Engineering, School of Engineering and Natural Sciences, Istanbul Medipol University, Istanbul 34810, Turkey
| | - Semih Macit
- Department of Biomedical Engineering, School of Engineering and Natural Sciences, Istanbul Medipol University, Istanbul 34810, Turkey
| | - Ummugulsum Yildiz
- Department of Biomedical Engineering, Erciyes University, Kayseri 38039, Turkey
- NanoThera Lab, ERFARMA-Drug Application and Research Center, Erciyes University, Kayseri 38280, Turkey
| | - Erhan Demirel
- Department of Biomedical Engineering, School of Engineering and Natural Sciences, Istanbul Medipol University, Istanbul 34810, Turkey
| | - Cansu Umran Tunç
- NanoThera Lab, ERFARMA-Drug Application and Research Center, Erciyes University, Kayseri 38280, Turkey
- Utah Center for Nanomedicine, University of Utah, Salt Lake City, Utah 84112, United States
| | - Halil Ulutabanca
- Department of Neurosurgery, Erciyes University Medical School, Kayseri 38030, Turkey
| | - Zuhal Hamurcu
- Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri 38030, Turkey
- GENKOK-Betül-Ziya Eren Genome and Stem Cell Center, Erciyes University, Kayseri 38280, Turkey
| | - Yasemin Yuksel Durmaz
- Department of Biomedical Engineering, School of Engineering and Natural Sciences, Istanbul Medipol University, Istanbul 34810, Turkey
- Research Institute of Health Science and Technologies (SABITA), Istanbul Medipol University, Istanbul 34810, Turkey
| | - Omer Aydin
- Department of Biomedical Engineering, Erciyes University, Kayseri 38039, Turkey
- NanoThera Lab, ERFARMA-Drug Application and Research Center, Erciyes University, Kayseri 38280, Turkey
- ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri 38039, Turkey
- ERKAM-Clinical-Engineering Research and Implementation Center, Erciyes University, Kayseri 38030, Turkey
| |
Collapse
|
16
|
Gupta G, Samuel VP, M RM, Rani B, Sasikumar Y, Nayak PP, Sudan P, Goyal K, Oliver BG, Chakraborty A, Dua K. Caspase-independent cell death in lung cancer: from mechanisms to clinical applications. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04149-0. [PMID: 40257494 DOI: 10.1007/s00210-025-04149-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 04/05/2025] [Indexed: 04/22/2025]
Abstract
Caspase-independent cell death (CICD) has recently become a very important mechanism in lung cancer, in particular, to overcome a critical failure in apoptotic cell death that is common to disease progression and treatment failures. The pathways involved in CICD span from necroptosis, ferroptosis, mitochondrial dysfunction, and autophagy-mediated cell death. Its potential therapeutic applications have been recently highlighted. Glutathione peroxidase 4 (GPX4) inhibition-driven ferroptosis has overcome drug resistance in non-small cell lung cancer (NSCLC). In addition, necroptosis involving RIPK1 and RIPK3 causes tumor cell death and modulation of immune responses in the tumor microenvironment (TME). Mitochondrial pathways are critical for CICD through modulation of metabolic and redox homeostasis. Ferroptosis is amplified by mitochondrial reactive oxygen species (ROS) and lipid peroxidation in lung cancer cells, and mitochondrial depolarization induces oxidative stress and leads to cell death. In addition, mitochondria-mediated autophagy, or mitophagy, results in the clearance of damaged organelles under stress conditions, while this function is also linked to CICD when dysregulated. The role of cell death through autophagy regulated by ATG proteins and PI3K/AKT/mTOR pathway is dual: to suppress tumor and to sensitize cells to therapy. A promising approach to enhancing therapeutic outcomes involves targeting mechanisms of CICD, including inducing ferroptosis by SLC7A11 inhibition, modulating mitochondrial ROS generation, or combining inhibition of autophagy with chemotherapy. Here, we review the molecular underpinnings of CICD, particularly on mitochondrial pathways and their potential to transform lung cancer treatment.
Collapse
Affiliation(s)
- Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Vijaya Paul Samuel
- Department of Anatomy, RAK College of Medicine, RAK Medical and Health Sciences University, Ras Al Khaimah, UAE
| | - Rekha M M
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Bindu Rani
- Department of Medicine, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - Y Sasikumar
- Department of CHEMISTRY, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Priya Priyadarshini Nayak
- Department of Medical Oncology IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, 751003, India
| | - Puneet Sudan
- Department of Pharmacy, Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to Be University), Clement Town, Dehradun, 248002, India
| | - Brian G Oliver
- Woolcock Institute of Medical Research, Macquarie University, Sydney, NSW, Australia
- School of Life Sciences, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Amlan Chakraborty
- Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Road, Manchester, M13 9PL, UK
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, VIC, 3800, Australia
| | - Kamal Dua
- Woolcock Institute of Medical Research, Macquarie University, Sydney, NSW, Australia.
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
| |
Collapse
|
17
|
Kamalanathan AS, Agarwal V, Talamini L, Muller S. Autophagy in myositis, a dysregulated pathway, and a target for therapy. Autoimmun Rev 2025; 24:103817. [PMID: 40262692 DOI: 10.1016/j.autrev.2025.103817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 03/27/2025] [Accepted: 04/14/2025] [Indexed: 04/24/2025]
Abstract
Corticosteroids and immunosuppressants are the mainstay of therapy for idiopathic inflammatory myopathies (IIMs). However, a significant therapeutic challenge extends beyond mitigating inflammation with these agents in achieving meaningful improvements in muscle strength and physical function, a goal that remains largely unmet. IIMs encompass a heterogeneous group of autoimmune disorders, including dermatomyositis, polymyositis, necrotizing autoimmune myopathy, inclusion body myositis, and others, characterized by chronic muscle inflammation, progressive weakness, and fatigue. The etiology of IIMs remains poorly understood, though potential contributors include environmental triggers (e.g., infections, medications, or injury) and genetic predisposition. To advance the development of novel therapeutic strategies, it is critical to elucidate the dysfunctional molecular and cellular pathways underlying IIM pathogenesis. Among these, dysregulated autophagy pathways have emerged as a promising target for therapeutic intervention. Specifically, impairments in lysosomal autophagy and mitophagy have been implicated in IIMs, and modulating these processes through targeted regulatory mechanisms may offer therapeutic benefits. This review provides a comprehensive synthesis of clinical and biological features of IIMs, the current diagnostic approaches and emerging biomarkers, evaluates the utility of existing biomarkers, and examines the relevance of animal models in IIM research. Furthermore, we explore the role of autophagic dysregulation in disease pathogenesis and provide a critical appraisal of current treatment modalities. Finally, we highlight emerging therapeutic targets and regulatory molecules under investigation, with a particular focus on autophagy modulation. Notably, autophagy inhibitors represent a novel and potentially transformative therapeutic avenue for patients with IIMs, offering hope for improved clinical outcomes.
Collapse
Affiliation(s)
- A S Kamalanathan
- Centre for BioSeparation Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - Vikas Agarwal
- Sanjay Gandhi Postgraduate Institute of Medical Sciences (SGPGIMS), Lucknow, Uttar Pradesh, India
| | - Laura Talamini
- CNRS and Strasbourg University Unit Biotechnology and Cell signalling/Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, France
| | - Sylviane Muller
- CNRS and Strasbourg University Unit Biotechnology and Cell signalling/Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, France; University of Strasbourg Institute for Advanced Study (USIAS), Strasbourg, France.
| |
Collapse
|
18
|
Glauser JSDO, Santana-Oliveira DA, Silva-Veiga FM, Fernandes-da-Silva A, Aguila MB, Souza-Mello V. Excessive dietary saturated fat or fructose and their combination (found in ultra-processed foods) impair mitochondrial dynamics markers and cause brown adipocyte whitening in adult mice. Nutrition 2025; 137:112805. [PMID: 40378644 DOI: 10.1016/j.nut.2025.112805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 03/05/2025] [Accepted: 04/07/2025] [Indexed: 05/19/2025]
Abstract
OBJECTIVE To investigate the effects of comparable dietary excess of fat or fructose and the combination of these two insults (mimicking ultra-processed foods) on interscapular brown adipose tissue (iBAT) whitening and markers of mitochondrial dynamics in adult male mice. METHODS Male C57BL/6 mice were randomly assigned into four groups according to the diet: control diet (C, following AIN-93M), high-fat diet (HF, 32% energy as lard), high-fructose diet (HFRU, 32% energy as fructose) or for high-fat/high-fructose diet (HF-HFRU, 32% as lard and 32% as fructose) for 12 weeks. Data were tested with one-way ANOVA and Dunnet T3 post-test (n=5 per analysis, significance level P < 0.05). RESULTS All diets caused insulin resistance and iBAT whitening, albeit with overweight only in the HF and HF-HFRU groups. Principal component analysis indicated that the HFRU scores loaded next to inflammation (Nlrp3) and adipogenesis markers (Pparg), and the HF diet influenced more a mitochondrial gene (Tomm20). However, iBAT whitening in all groups was associated with deficits in mitochondrial dynamics (Ppargc1a, Dnml1, and Pink1), vascularization (Vegfa), and thermogenic markers (Bmp8b, and Ucp1). CONCLUSION Similar increases in dietary saturated fat or fructose (32% as energy) and the combination of these two insults (32% / 32%) caused insulin resistance and brown adipocyte dysfunction (whitening) in adult mice after 12 weeks independent of being overweight. In comparison, the PC scores of the HFRU groups were closer to the HF-HFRU group than the HF group, implying a worse outcome and highlighting the importance of limiting saturated fat and fructose intake from ultra-processed foods.
Collapse
Affiliation(s)
- Jade Sancha de Oliveira Glauser
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Daiana Araujo Santana-Oliveira
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Flávia Maria Silva-Veiga
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Aline Fernandes-da-Silva
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcia Barbosa Aguila
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vanessa Souza-Mello
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
19
|
Tayir M, Wang YW, Chu T, Wang XL, Fan YQ, Cao L, Chen YH, Wu DD. The function of necroptosis in liver cancer. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167828. [PMID: 40216370 DOI: 10.1016/j.bbadis.2025.167828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 02/20/2025] [Accepted: 03/30/2025] [Indexed: 04/26/2025]
Abstract
Liver cancer is one of the most lethal cancers, and apoptosis resistance is a major obstacle contributing to chemotherapy failure in liver cancer treatment. Inducing cancer cell death by bypassing the apoptotic pathway is considered a promising approach to overcome this problem. Necroptosis is a non-caspase-dependent regulated mode of cell death mainly mediated by receptor-interacting protein kinase 1 (RIPK1), RIPK3, and mixed lineage kinase domain-like (MLKL) protein, and the utilization of necroptosis for treating hepatocellular carcinoma (HCC) also offers a new hope for addressing liver cancer in the clinic. In this paper, the role of necroptosis in HCC as well as the effect on differentiation of liver cancer are reviewed. We also comparatively analyze the relationship among necroptosis, apoptosis, and necrosis, as well as summarize the characteristics and functions of key proteins involved in this pathway. The bidirectional regulation of necroptosis and the mitochondrial machinery within this pathway deserve attention.
Collapse
Affiliation(s)
- Mukaddas Tayir
- Department of Stomatology, Huaihe Hospital of Henan University, School of Stomatology, Henan University, Kaifeng, Henan 475004, China
| | - Yan-Wen Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China
| | - Ti Chu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China
| | - Xue-Li Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China
| | - Yong-Qi Fan
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China
| | - Lei Cao
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China
| | - Yu-Hang Chen
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China
| | - Dong-Dong Wu
- Department of Stomatology, Huaihe Hospital of Henan University, School of Stomatology, Henan University, Kaifeng, Henan 475004, China; Kaifeng Key Laboratory of Periodontal Tissue Engineering, School of Stomatology, Henan University, Kaifeng, Henan 475004, China.
| |
Collapse
|
20
|
Tang Z, Xue Z, Liu X, Zhang Y, Zhao J, Liu J, Zhang L, Guo Q, Feng B, Wang J, Zhang D, Li X. Inhibition of hypoxic exosomal miR-423-3p decreases glioma progression by restricting autophagy in astrocytes. Cell Death Dis 2025; 16:265. [PMID: 40199864 PMCID: PMC11978802 DOI: 10.1038/s41419-025-07576-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 02/23/2025] [Accepted: 03/18/2025] [Indexed: 04/10/2025]
Abstract
The tumor microenvironment (TME) of gliomas comprises glioma cells and surrounding cells, such as astrocytes, macrophages, T cells, and neurons. In the TME, glioma cells can activate normal human astrocytes (NHAs) through the secretion of exosomes and the activation of astrocytes can further improve the progression of glioma, leading to a poor prognosis for patients. However, the molecular mechanisms underlying NHAs activation by gliomas remain largely unknown. It this study, glioma-derived exosomes (GDEs) play an important role in the modulation of autophagy and activation of NHAs. Compared with normoxic GDEs, hypoxic glioma-derived exosomes (H-GDEs) further improved autophagy and activation of astrocytes, which strongly promoted the progression of glioma cells. In an miRNA array between two types of exosomes from gliomas, miR-423-3p was highly expressed in H-GDEs and played an important role in autophagy, resulting in the activation of NHAs. The mechanism by which hypoxic glioma cells react with NHAs to create an immunosuppressive microenvironment was identified and 15d-PGJ2 was established as an effective inhibitor of miR-423-3p to suppress NHAs activation. These findings provide new insights into the diagnosis and treatment of gliomas by targeting autophagy and miR-423-3p expression.
Collapse
Affiliation(s)
- Ziyi Tang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China
| | - Zhiwei Xue
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China
| | - Xuchen Liu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China
| | - Yan Zhang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China
| | - Jiangli Zhao
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China
| | - Junzhi Liu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China
| | - Lin Zhang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Qindong Guo
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China
| | - Bowen Feng
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China
| | - Jiwei Wang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China.
| | - Di Zhang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China.
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, China.
| |
Collapse
|
21
|
Ma Z, Cen Y, Xun W, Mou C, Yu J, Hu Y, Liu C, Sun J, Bi R, Qiu Y, Ding M, Jin L. Exercise enhances cardiomyocyte mitochondrial homeostasis to alleviate left ventricular dysfunction in pressure overload induced remodelling. Sci Rep 2025; 15:11698. [PMID: 40188200 PMCID: PMC11972341 DOI: 10.1038/s41598-025-95637-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 03/24/2025] [Indexed: 04/07/2025] Open
Abstract
This study aims to explore how exercise enhances mitochondrial regulation and mitigates pathological cardiac hypertrophy. Rat groups were assigned as the control group (CN, n = 8), sham group (sham, n = 8), model group (SC, n = 16) and exercise group (SE, n = 20). A bioinformatics analysis was conducted to uncover the underlying mechanisms.H9C2 cells were divided into: the Ang II 0 h group (CON), Ang II 48 h group (Ang II), Ang II 48 h + sh-control group (sh-GFP + Ang II), Ang II 48 h + sh-ndufb10 group (sh-ndufb10 + Ang II), Ang II 48 h + overexpressedndufb10 control group (Ad-GFP + Ang II) and Ang II 48 h + over-expressedndufb10group (Ad-ndufb10 + Ang II). Mitochondrial function was measured. mRNA and protein expression were assessed by qPCR or western blot analysis respectively. In the SC group, a significant increase was observed in cardiomyocyte diameter, cardiac function, autophagy, and apoptosis. After 8 weeks of swimming exercise, there was a substantial reduction in cardiomyocyte diameter, an improvement in cardiac function, a mitigation of mitochondrial fission and autophagy. Ndufb10 was markedly enriched in oxidative phosphorylation and downregulated in the SC group, while it was upregulated in the SE group. In the sh-ndufb10 group, mitochondrial fusion was suppressed; fission and autophagy were further facilitated; mitochondrial membrane potential, mPTP, and ROS levels increased; and TUNEL positive nuclei and apoptosis-related proteins showed significant upregulation. Overexpression of ndufb10 reversed pathological hypertrophy, mitochondrial autophagy, mitochondrial dysfunction, and cardiomyocyte apoptosis in vitro. Swimming exercise improves mitochondrial abnormalities and reduces cardiomyocyte hypertrophy through regulation of the ndufb10 in left ventricular hypertrophy.
Collapse
Affiliation(s)
- Zhichao Ma
- School of Physical Education, Wuhan Business University, Wuhan, 430056, China.
- College of Health Science, Wuhan Sports University, Wuhan, 430079, China.
- Equine Science Research and Horse Doping Control Laboratory, Wuhan Business University, Wuhan, 430056, China.
| | - Yanling Cen
- School of Physical Education, Wuhan Business University, Wuhan, 430056, China
- College of Health Science, Wuhan Sports University, Wuhan, 430079, China
- Hubei Exercise Training and Monitoring Key Laboratory, Wuhan Sports University, Wuhan, 430079, China
| | - Weiwei Xun
- College of Health Science, Wuhan Sports University, Wuhan, 430079, China
- Hubei Exercise Training and Monitoring Key Laboratory, Wuhan Sports University, Wuhan, 430079, China
| | - Caiying Mou
- College of Health Science, Wuhan Sports University, Wuhan, 430079, China
- Hubei Exercise Training and Monitoring Key Laboratory, Wuhan Sports University, Wuhan, 430079, China
| | - Junwen Yu
- Aquinas International Academy, Ontario, CA, 90623, USA
| | - Yarui Hu
- Chiko Sports Institute, Sichuan University of Science and Technology, Meishan, 620000, China
| | - Chen Liu
- College of Health Science, Wuhan Sports University, Wuhan, 430079, China
- Hubei Exercise Training and Monitoring Key Laboratory, Wuhan Sports University, Wuhan, 430079, China
| | - Jun Sun
- College of Health Science, Wuhan Sports University, Wuhan, 430079, China
- Hubei Exercise Training and Monitoring Key Laboratory, Wuhan Sports University, Wuhan, 430079, China
| | - Rui Bi
- College of Health Science, Wuhan Sports University, Wuhan, 430079, China
- Hubei Exercise Training and Monitoring Key Laboratory, Wuhan Sports University, Wuhan, 430079, China
| | - Yanli Qiu
- College of Health Science, Wuhan Sports University, Wuhan, 430079, China
- Hubei Exercise Training and Monitoring Key Laboratory, Wuhan Sports University, Wuhan, 430079, China
| | - Mingchao Ding
- College of Health Science, Wuhan Sports University, Wuhan, 430079, China
- Hubei Exercise Training and Monitoring Key Laboratory, Wuhan Sports University, Wuhan, 430079, China
| | - Li Jin
- College of Health Science, Wuhan Sports University, Wuhan, 430079, China.
- Hubei Exercise Training and Monitoring Key Laboratory, Wuhan Sports University, Wuhan, 430079, China.
| |
Collapse
|
22
|
Chen Y, Zhang D, Li J, Sun Y, Wang J, Xi L. SNS‑032 combined with decitabine induces caspase‑3/gasdermin E‑dependent pyroptosis in breast cancer cells. Oncol Lett 2025; 29:202. [PMID: 40070781 PMCID: PMC11894506 DOI: 10.3892/ol.2025.14948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 01/13/2025] [Indexed: 03/14/2025] Open
Abstract
SNS-032 is a synthetic compound that specifically inhibits cyclin-dependent kinases 2, 7 and 9. Its primary anticancer activity involves cell cycle arrest, which prevents tumor cell growth. However, there are limited reports on whether SNS-032 induces pyroptosis, a novel inflammation-mediated programmed cell death pathway in breast cancer (BC). The present study demonstrated that SNS-032 treatment decreased cell viability by inducing pyroptosis in BC cells. Typical morphological indications of pyroptosis were observed, including cell swelling and destruction of cell membrane integrity, leading the release of adenosine 5'-triphosphate and lactate dehydrogenase. Furthermore, the expression of caspase-3, the N terminus of gasdermin E (GSDME) and B-cell lymphoma-2 (BCL-2)-associated X protein increased, whereas expression of BCL-2 decreased. In addition, Z-DEVD-FMK, a specific caspase-3 inhibitor, markedly alleviated pyroptosis triggered by SNS-032. These findings suggested that SNS-032 induced caspase-3/GSDME-dependent pyroptosis. Furthermore, the present study demonstrated that decitabine (DAC), a DNA methyltransferase inhibitor, upregulated the expression of GSDME protein and enhanced SNS-032-induced caspase-3/GSDME-dependent pyroptosis in BC cells. In conclusion, these results suggest that caspase-3/GSDME-induced pyroptosis can be facilitated by SNS-032 treatment in BC cells, and DAC has the potential to enhance SNS-032-induced pyroptosis by increasing GSDME expression. This mechanistic insight indicates that SNS-032 is a promising therapeutic agent for BC treatment.
Collapse
Affiliation(s)
- Yuxin Chen
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Danya Zhang
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Jie Li
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yue Sun
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Jing Wang
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Ling Xi
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
23
|
Guo B, Gu J, Zhuang T, Zhang J, Fan C, Li Y, Zhao M, Chen R, Wang R, Kong Y, Xu S, Gao W, Liang L, Yu H, Han T. MicroRNA-126: From biology to therapeutics. Biomed Pharmacother 2025; 185:117953. [PMID: 40036996 DOI: 10.1016/j.biopha.2025.117953] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/22/2025] [Accepted: 02/27/2025] [Indexed: 03/06/2025] Open
Abstract
MicroRNA-126 (miR-126) has emerged as one of the most extensively studied microRNAs in the context of human diseases, particularly in vascular disorders and cancer. Its high degree of conservation across vertebrates underscores its evolutionary significance and essential functional roles. Extensive research has been devoted to elucidating the molecular mechanisms through which miR-126 modulates key physiological and pathological processes, including angiogenesis, immune response, inflammation, tumor growth, and metastasis. Furthermore, miR-126 plays a causal role in the pathogenesis of various diseases, serving as potential biomarkers for disease prediction, diagnosis, prognosis and drug response, as well as a promising therapeutic target. In this review, we synthesize findings from 283 articles, focusing on the roles of miR-126 in critical biological processes such as cell development, survival, cycle regulation, proliferation, migration, invasion, communication, and metabolism. Additionally, miR-126 represents a promising candidate for miRNA-based therapeutic strategies. A comprehensive understanding and evaluation of miR-126 are crucial for advancing its clinical applications and therapeutic potential.
Collapse
Affiliation(s)
- Bei Guo
- Department of Metabolism and Endocrinology, General Hospital of Northern Theater Command, Shenyang, China
| | - Jia Gu
- Department of Otolaryngology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Tongtian Zhuang
- Department of Dermatology, Air Force Hospital of Northern Theater Command, Shenyang, China
| | - Jingbin Zhang
- Department of Metabolism and Endocrinology, General Hospital of Northern Theater Command, Shenyang, China
| | - Chunyang Fan
- Department of Metabolism and Endocrinology, General Hospital of Northern Theater Command, Shenyang, China
| | - Yiyao Li
- Department of Metabolism and Endocrinology, General Hospital of Northern Theater Command, Shenyang, China
| | - Mengdi Zhao
- Department of Metabolism and Endocrinology, General Hospital of Northern Theater Command, Shenyang, China
| | - Ruoran Chen
- Department of Metabolism and Endocrinology, General Hospital of Northern Theater Command, Shenyang, China
| | - Rui Wang
- Department of Metabolism and Endocrinology, General Hospital of Northern Theater Command, Shenyang, China
| | - Yuan Kong
- Department of Metabolism and Endocrinology, General Hospital of Northern Theater Command, Shenyang, China
| | - Shuang Xu
- Department of Metabolism and Endocrinology, General Hospital of Northern Theater Command, Shenyang, China
| | - Wei Gao
- Department of Metabolism and Endocrinology, General Hospital of Northern Theater Command, Shenyang, China
| | - Linlang Liang
- Department of Metabolism and Endocrinology, General Hospital of Northern Theater Command, Shenyang, China
| | - Hao Yu
- Department of Metabolism and Endocrinology, General Hospital of Northern Theater Command, Shenyang, China.
| | - Tao Han
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
24
|
Wang R, Wang T, Chen Z, Jiang J, Du Y, Yuan H, Pan Y, Wang Y. Bioactive materials from berberine-treated human bone marrow mesenchymal stem cells accelerate tooth extraction socket healing through the jaw vascular unit. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1025-1041. [PMID: 39825206 DOI: 10.1007/s11427-024-2745-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 09/25/2024] [Indexed: 01/20/2025]
Abstract
Delayed tooth extraction socket (TES) healing can cause failure of subsequent oral implantation and increase socioeconomic burden on patients. Excessive amounts of M1 macrophages, apoptotic neutrophils (ANs), and neutrophil extracellular traps (NETs) impair alveolar bone regeneration during TES healing. In the present study, we first discovered that conditioned medium (CM) collected from berberine-treated human bone marrow mesenchymal stem cells (BBR-HB-CM) accelerated TES healing. BBR-HB-CM contained bioactive materials that promoted the polarization of macrophages from M1 to M2, impeded the formation of ANs and NETs, and modulated M2 macrophage efferocytosis in vivo and in vitro. Mechanistically, BBR-HB-CM promoted bone formation by inhibiting macrophage-myofibroblast transition and reprogrammed macrophage polarization through p85/AKT/mTOR pathway-dependent autophagy. The 3-methyladenine abolished the therapeutic effects of BBR-HB-CM. Further studies revealed that BBR-HB-CM accelerated TES healing in rats with type 2 diabetes mellitus. Overall, our results demonstrated that BBR-HB-CM had high potential to promote rapid TES healing.
Collapse
Affiliation(s)
- Ruyu Wang
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University; State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases; Jiangsu Province Engineering Research Centre of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, 210029, China
- Department of Stomatology, Chongzhou People's Hospital, Chengdu, 611230, China
| | - Tianxiao Wang
- Medical Basic Research Innovation Centre for Cardiovascular and Cerebrovascular Diseases, Ministry of Education; International Joint Laboratory for Drug Target of Critical Illnesses; School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Ziyu Chen
- Medical Basic Research Innovation Centre for Cardiovascular and Cerebrovascular Diseases, Ministry of Education; International Joint Laboratory for Drug Target of Critical Illnesses; School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Jiandong Jiang
- Medical Basic Research Innovation Centre for Cardiovascular and Cerebrovascular Diseases, Ministry of Education; International Joint Laboratory for Drug Target of Critical Illnesses; School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China.
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| | - Yifei Du
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University; State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases; Jiangsu Province Engineering Research Centre of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Hua Yuan
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University; State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases; Jiangsu Province Engineering Research Centre of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Yongchu Pan
- Department of Orthodontic, The Affiliated Stomatological Hospital of Nanjing Medical University; State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases; Jiangsu Province Engineering Research Centre of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Yuli Wang
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University; State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases; Jiangsu Province Engineering Research Centre of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, 210029, China.
- Medical Basic Research Innovation Centre for Cardiovascular and Cerebrovascular Diseases, Ministry of Education; International Joint Laboratory for Drug Target of Critical Illnesses; School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
25
|
Dagher DM, Zaghloul MS, Suddek GM. Modulation of AMPK/mTOR Autophagic Pathway Using Dapagliflozin Protects Against Cadmium-Induced Testicular and Renal Injury in Rats. J Biochem Mol Toxicol 2025; 39:e70257. [PMID: 40233265 DOI: 10.1002/jbt.70257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 02/03/2025] [Accepted: 03/31/2025] [Indexed: 04/17/2025]
Abstract
Cadmium is a widely distributed heavy metal found in the environment that poses serious hazards to human health. Dapagliflozin (DAPA), a sodium-glucose co-transporter 2 (SGLT-2) inhibitor, exhibited antioxidant, antiapoptotic, and anti-inflammatory properties. Our data assessed the effect of DAPA against Cd-triggered renal and testicular impairment in rats, as well as the underlying mechanisms. Cd (30 mg/kg) and DAPA (5 and 10 mg/kg) were administrated by oral gavage to rats and continued for 21 days. DAPA attenuated Cd-triggered renal and testicular injury as shown by diminishing serum creatinine, BUN, and urinary total protein concentration in addition to increasing creatinine clearance, urinary creatinine, and serum testosterone. Moreover, it diminished renal and testicular histopathological alterations induced by Cd. DAPA stimulated the impaired autophagy flux as seen by significantly elevating the p-AMPK/total AMPK, decreasing p-mTOR/total mTOR ratios, and diminishing p62 & LC3 protein levels. Additionally, DAPA significantly lowered MDA content, increased GSH level and SOD activity. Moreover, it augmented the cytoprotective Nrf2/HO-1 signaling pathway. Furthermore, it attenuated renal and testicular apoptotic cell death via decreasing caspase-3 expression. Conclusion: Boosting autophagic events and combating oxidative stress and apoptosis by DAPA were engaged in alleviating Cd-induced renal and testicular impairment. This was accomplished by modulating the AMPK/mTOR and enhancing the Nrf2/HO-1 pathways.
Collapse
Affiliation(s)
- Doha M Dagher
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Marwa S Zaghloul
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura National University, Gamasa, Egypt
| | - Ghada M Suddek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
26
|
Yang Y, Ma C, Wang Y, Tian J, Li B, Zhao J. Pectin-coated Malvidin-3-O-galactoside attenuates silica-induced pulmonary fibrosis by promoting mitochondrial autophagy and inhibiting cell apoptosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156566. [PMID: 40023066 DOI: 10.1016/j.phymed.2025.156566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 02/11/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND Blueberries are a rich source of anthocyanins, which have been established to have multiple beneficial properties. However, the structure of anthocyanin monomers is unstable and their bioavailability is low. To date, whereas functional studies on anthocyanins have focused mainly on the effects of their monomers on liver and kidney, few have examined the interventional effects on pulmonary fibrosis. PURPOSE In this study, we combined malvidin-3-O-galactoside (M3G)1 derived from blueberries with pectin (PEC)2 to form an anthocyanin-pectin complex (M3G-PEC),3 the anti-fibrotic effects of which were examined by administering to mice with modeled pulmonary fibrosis induced by silica particles (SP).4 METHODS: To evaluate the therapeutic effects and mechanisms of action of M3G-PEC with respect to the progression of pulmonary fibrosis, we measured autophagy- and apoptosis-related indices in C57BL/6 mice and mouse alveolar macrophage cell line (MH-S).5 RESULTS: The results of in vivo and in vitro studies revealed that M3G-PEC can alleviate the degree of pulmonary fibrosis, enhances the expression of Microtubule-associated protein light chain 3 (LC3),6 PTEN-inducible putative kinase 1 (PINK1),7 Parkin and B-cell lymphoma-2 (BCL-2),8 and causes the down-regulation of Caspase-3, P62, p-mammalian target of rapamyein (p-mTOR),9 phosphorylated protein kinase B (p-Akt)10 and Bax. And then, M3G-PEC contributes to maintaining a steady mitochondrial membrane potential and reduces the release of cytochrome c (Cyt-C)11 in cells. CONCLUSION Collectively, these findings indicate that M3G-PEC can preserve the bioactivity of anthocyanins and effectively enhance their bioavailability. Moreover, by regulating the BECN-1/Akt/mTOR pathway, M3G-PEC can influence the progression of silica-induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Yihan Yang
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning 110866, China.
| | - Chuang Ma
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning 110866, China.
| | - Yining Wang
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning 110866, China.
| | - Jinlong Tian
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning 110866, China.
| | - Bin Li
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning 110866, China.
| | - Jin Zhao
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning 110866, China.
| |
Collapse
|
27
|
Kuzminsky I, Ghanim M. Immunity responses as checkpoints for efficient transmission of begomoviruses by whiteflies. Virology 2025; 605:110462. [PMID: 40020542 DOI: 10.1016/j.virol.2025.110462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/05/2025] [Accepted: 02/20/2025] [Indexed: 03/03/2025]
Abstract
Begomoviruses are a group of single stranded DNA plant viruses exclusively transmitted by the sweet potato whitefly Bemisia tabaci in a persistent, circulative manner. After acquisition from plant phloem, this group of viruses circulate and are retained within the whitefly, interacting with tissues, cells and molecular pathways for maintaining the safety of the infective intact virions, by exploiting cellular mechanisms and avoiding degradation by the insect immune responses. During retention, the virions are internalized in the midgut cells, exit and spend hours-days in the hemolymph and cross into salivary gland cells, before transmission. Destroying this group of viruses by the insect immune system seems inefficient for the most part, by examining their very efficient transmission. Thus, within the various sites along the transmission pathway especially in the midgut, it is thought that the immune system with its various layers is activated for avoiding the damage caused by the viruses on one hand, and for ensuring their safe circulation and transmission on the other hand. Begomoviruses have evolved mechanisms for counteracting and exploiting the activated immune system for their safe translocation within the whitefly. In this review, we discuss the various levels of immunity activated against begomoviruses in B. tabaci, taking other pathogen-vector systems as examples and reflecting relevant components on the interactions between B. tabaci and Begomoviruses.
Collapse
Affiliation(s)
- Ilana Kuzminsky
- Department of Entomology, Volcani Center, Rishon LeZion, 7505101, Israel; Department of Agroecology and Plant Health, Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, 7610001, Israel
| | - Murad Ghanim
- Department of Entomology, Volcani Center, Rishon LeZion, 7505101, Israel.
| |
Collapse
|
28
|
Wang C, Liu X, Zhao Y, Liao S, Zhang J, Huang Y, Shi Y, Li L, Pan Q, Wu J, Wang Y. AMPK activation by hepatitis E virus infection inhibits viral replication through attenuation of autophagosomes and promotion of innate immunity. Cell Mol Life Sci 2025; 82:111. [PMID: 40074929 PMCID: PMC11904043 DOI: 10.1007/s00018-025-05634-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 01/26/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025]
Abstract
Hepatitis E virus (HEV) infection is generally asymptomatic or leads to acute and self-limiting hepatitis. The mechanisms orchestrating such an infection course remain to be elucidated. AMP-activated protein kinase (AMPK) is a pivotal cellular sensor for maintaining metabolic homeostasis. Here, we show that AMPK is activated in response to HEV infection and is associated with mitochondrial damage and ATP deficiency. AMPK activation, in turn, inhibits HEV replication. Mechanistic studies reveal that AMPK activation triggers the expression of interferon (IFN)-stimulated genes that possess antiviral properties. In parallel, AMPK inhibits autophagosome accumulation to exert antiviral effects. Interestingly, AMPK activation also suppresses the inflammatory response triggered by HEV infection. Consistently, AMPK activation simultaneously exerts anti-inflammatory and antiviral effects in a coculture system of HEV-infected liver cells with macrophages. These findings pave the way for the development of AMPK-targeted therapeutics to treat hepatitis E.
Collapse
Affiliation(s)
- Chunling Wang
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China
- Institute of Reproductive Health/NHC Key Laboratory of Birth Defects Prevention, Henan Academy of Innovations in Medical Science, Zhengzhou, China
| | - Xiaoman Liu
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yao Zhao
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Shumin Liao
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Jiayue Zhang
- School of Pharmacy, Jiangsu Food & Pharmaceutical Science College, Huaian, Jiangsu, China
| | - Yanhong Huang
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yue Shi
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Liang Li
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Qiuwei Pan
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, 3015CE, The Netherlands.
| | - Jian Wu
- Department of Clinical Laboratory, The Affiliated Suzhou Hospital of Nnjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, 242 Guangji Rd, Suzhou, Jiangsu, 215008, China.
| | - Yijin Wang
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China.
| |
Collapse
|
29
|
Zhao N, Xu A, Yang J, Zhao J, Xie J, Li B, Duan J, Cao G. Triclosan Caused Oocyte Meiotic Arrest by Modulating Oxidative Stress, Organelle Dysfunctions, Autophagy, and Apoptosis in Pigs. Animals (Basel) 2025; 15:802. [PMID: 40150331 PMCID: PMC11939393 DOI: 10.3390/ani15060802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/05/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
Triclosan (TCS) is a highly effective broad-spectrum antibacterial agent; however, the specific roles of TCS in oocyte maturation remain poorly understood. This research investigated the influence of TCS on biologically active processes during the in vitro maturation of porcine oocytes. Our results demonstrated that TCS significantly decreased the maturation rate of porcine oocytes in a concentration-dependent manner and impaired cumulus expansion. These detrimental effects were mediated by the disruption of mitochondrial function and distribution, leading to oxidative stress characterized by an accumulation of reactive oxygen species (ROS), a decrease in the expression of the antioxidant enzymes SOD2 and GSH, reduced ATP production, and a loss of mitochondrial membrane potential (ΔΨm). We also observed interference with endoplasmic reticulum (ER) distribution, disturbances in Ca2+ homeostasis, and fluctuations in ER stress, as evidenced by reduced expression of ER stress-related proteins. Furthermore, TCS exposure induced autophagy, as indicated by the levels of SQSTM1 (P62) and LC3-II. Additionally, TCS increased apoptosis rates, corresponding with a downregulation of Bcl-2 expression. Collectively, our findings suggest that exposure to TCS can impair cytoplasmic function, thereby affecting oocyte quality.
Collapse
Affiliation(s)
- Ning Zhao
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China; (N.Z.); (A.X.); (J.Y.); (J.Z.); (J.X.); (B.L.)
- Shanxi Key Laboratory of Animal Genetics Resource Utilization and Breeding, Jinzhong 030801, China
| | - Anli Xu
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China; (N.Z.); (A.X.); (J.Y.); (J.Z.); (J.X.); (B.L.)
- Shanxi Key Laboratory of Animal Genetics Resource Utilization and Breeding, Jinzhong 030801, China
| | - Jingxian Yang
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China; (N.Z.); (A.X.); (J.Y.); (J.Z.); (J.X.); (B.L.)
- Shanxi Key Laboratory of Animal Genetics Resource Utilization and Breeding, Jinzhong 030801, China
| | - Jianan Zhao
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China; (N.Z.); (A.X.); (J.Y.); (J.Z.); (J.X.); (B.L.)
- Shanxi Key Laboratory of Animal Genetics Resource Utilization and Breeding, Jinzhong 030801, China
| | - Junhao Xie
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China; (N.Z.); (A.X.); (J.Y.); (J.Z.); (J.X.); (B.L.)
- Shanxi Key Laboratory of Animal Genetics Resource Utilization and Breeding, Jinzhong 030801, China
| | - Bugao Li
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China; (N.Z.); (A.X.); (J.Y.); (J.Z.); (J.X.); (B.L.)
- Shanxi Key Laboratory of Animal Genetics Resource Utilization and Breeding, Jinzhong 030801, China
| | - Jiaxin Duan
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China; (N.Z.); (A.X.); (J.Y.); (J.Z.); (J.X.); (B.L.)
- Shanxi Key Laboratory of Animal Genetics Resource Utilization and Breeding, Jinzhong 030801, China
| | - Guoqing Cao
- College of Animal Science, Shanxi Agricultural University, Jinzhong 030801, China; (N.Z.); (A.X.); (J.Y.); (J.Z.); (J.X.); (B.L.)
- Shanxi Key Laboratory of Animal Genetics Resource Utilization and Breeding, Jinzhong 030801, China
| |
Collapse
|
30
|
Du B, Fu Q, Yang Q, Yang Y, Li R, Yang X, Yang Q, Li S, Tian J, Liu H. Different types of cell death and their interactions in myocardial ischemia-reperfusion injury. Cell Death Discov 2025; 11:87. [PMID: 40044643 PMCID: PMC11883039 DOI: 10.1038/s41420-025-02372-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 01/10/2025] [Accepted: 02/21/2025] [Indexed: 03/09/2025] Open
Abstract
Myocardial ischemia-reperfusion (I/R) injury is a multifaceted process observed in patients with coronary artery disease when blood flow is restored to the heart tissue following ischemia-induced damage. Cardiomyocyte cell death, particularly through apoptosis, necroptosis, autophagy, pyroptosis, and ferroptosis, is pivotal in myocardial I/R injury. Preventing cell death during the process of I/R is vital for improving ischemic cardiomyopathy. These multiple forms of cell death can occur simultaneously, interact with each other, and contribute to the complexity of myocardial I/R injury. In this review, we aim to provide a comprehensive summary of the key molecular mechanisms and regulatory patterns involved in these five types of cell death in myocardial I/R injury. We will also discuss the crosstalk and intricate interactions among these mechanisms, highlighting the interplay between different types of cell death. Furthermore, we will explore specific molecules or targets that participate in different cell death pathways and elucidate their mechanisms of action. It is important to note that manipulating the molecules or targets involved in distinct cell death processes may have a significant impact on reducing myocardial I/R injury. By enhancing researchers' understanding of the mechanisms and interactions among different types of cell death in myocardial I/R injury, this review aims to pave the way for the development of novel interventions for cardio-protection in patients affected by myocardial I/R injury.
Collapse
Affiliation(s)
- Bingxin Du
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qiang Fu
- Department of Chinese Formulae, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qin Yang
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yeying Yang
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Rui Li
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xu Yang
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qingrong Yang
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shuo Li
- Heilongjiang Provincial Key Laboratory of Panvascular Disease, Harbin, China
| | - Jinwei Tian
- Heilongjiang Provincial Key Laboratory of Panvascular Disease, Harbin, China.
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Huibin Liu
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
- Heilongjiang Provincial Key Laboratory of Panvascular Disease, Harbin, China.
| |
Collapse
|
31
|
Wang X, Xu L, Wu Z, Lou L, Xia C, Miao H, Dai J, Fei W, Wang J. Exosomes of stem cells: a potential frontier in the treatment of osteoarthritis. PRECISION CLINICAL MEDICINE 2025; 8:pbae032. [PMID: 39781279 PMCID: PMC11705996 DOI: 10.1093/pcmedi/pbae032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/18/2024] [Accepted: 11/25/2024] [Indexed: 01/12/2025] Open
Abstract
The aging population has led to a global issue of osteoarthritis (OA), which not only impacts the quality of life for patients but also poses a significant economic burden on society. While biotherapy offers hope for OA treatment, currently available treatments are unable to delay or prevent the onset or progression of OA. Recent studies have shown that as nanoscale bioactive substances that mediate cell communication, exosomes from stem cell sources have led to some breakthroughs in the treatment of OA and have important clinical significance. This paper summarizes the mechanism and function of stem cell exosomes in delaying OA and looks forward to the development prospects and challenges of exosomes.
Collapse
Affiliation(s)
- Xiaofei Wang
- The Graduate School, Dalian Medical University, Dalian 116044, China
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Lei Xu
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Zhimin Wu
- The Graduate School, Dalian Medical University, Dalian 116044, China
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Linbing Lou
- The Graduate School, Dalian Medical University, Dalian 116044, China
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Cunyi Xia
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Haixiang Miao
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Jihang Dai
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Wenyong Fei
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Jingcheng Wang
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| |
Collapse
|
32
|
Luo PY, Zou JR, Chen T, Zou J, Li W, Chen Q, Cheng L, Zheng LY, Qian B. Autophagy in erectile dysfunction: focusing on apoptosis and fibrosis. Asian J Androl 2025; 27:166-176. [PMID: 39028624 PMCID: PMC11949458 DOI: 10.4103/aja202433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 04/22/2024] [Indexed: 07/21/2024] Open
Abstract
ABSTRACT In most types of erectile dysfunction, particularly in advanced stages, typical pathological features observed are reduced parenchymal cells coupled with increased tissue fibrosis. However, the current treatment methods have shown limited success in reversing these pathologic changes. Recent research has revealed that changes in autophagy levels, along with alterations in apoptosis and fibrosis-related proteins, are linked to the progression of erectile dysfunction, suggesting a significant association. Autophagy, known to significantly affect cell fate and tissue fibrosis, is currently being explored as a potential treatment modality for erectile dysfunction. However, these present studies are still in their nascent stage, and there are limited experimental data available. This review analyzes erectile dysfunction from a pathological perspective. It provides an in-depth overview of how autophagy is involved in the apoptotic processes of smooth muscle and endothelial cells and its role in the fibrotic processes occurring in the cavernosum. This study aimed to develop a theoretical framework for the potential effectiveness of autophagy in preventing and treating erectile dysfunction, thus encouraging further investigation among researchers in this area.
Collapse
Affiliation(s)
- Pei-Yue Luo
- The First Clinical College, Gannan Medical University, Ganzhou 341000, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou 341000, China
| | - Jun-Rong Zou
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou 341000, China
| | - Tao Chen
- The First Clinical College, Gannan Medical University, Ganzhou 341000, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou 341000, China
| | - Jun Zou
- The First Clinical College, Gannan Medical University, Ganzhou 341000, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou 341000, China
| | - Wei Li
- The First Clinical College, Gannan Medical University, Ganzhou 341000, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou 341000, China
| | - Qi Chen
- The First Clinical College, Gannan Medical University, Ganzhou 341000, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou 341000, China
| | - Le Cheng
- The First Clinical College, Gannan Medical University, Ganzhou 341000, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou 341000, China
| | - Li-Ying Zheng
- Department of Graduate, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Biao Qian
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou 341000, China
| |
Collapse
|
33
|
Chen P, Chen Y, Sharma A, Gonzalez-Carmona Maria A, Schmidt-Wolf IGH. Inhibition of ERO1L induces autophagy and apoptosis via endoplasmic reticulum stress in colorectal cancer. Cell Signal 2025; 127:111560. [PMID: 39657838 DOI: 10.1016/j.cellsig.2024.111560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 12/03/2024] [Accepted: 12/06/2024] [Indexed: 12/12/2024]
Abstract
Colorectal cancer (CRC) is one of the most common types of cancer with high incidence and mortality. Endoplasmic reticulum oxidoreductase 1 alpha (ERO1L) is overexpressed in CRC. This study aims to explore the role of ERO1L in CRC progression and evaluate the anti-tumor efficacy of the combination treatment of ERO1L inhibition with endoplasmic reticulum (ER) stress-inducing therapies. Herein, we found that ERO1L was elevated in CRC cell lines and patients. ER stress upregulated the expression of ERO1L, and ERO1L deficiency induced ER stress in CRC. ERO1L knockdown increased the susceptibility of CRC cells to ER stress. ERO1L contributed to the malignant phenotypes of CRC cells. Inhibition of ERO1L induced autophagy and caspase-dependent apoptosis by the induction of ER stress in CRC cells. Mechanically, the ERK1/2 pathway was involved in ERO1L knockdown-mediated apoptosis and autophagy. Combination treatment of ERO1L inhibition with ER stress-inducing agents, such as unfolded protein response (UPR)-targeting inhibitors and proteasome inhibitors, demonstrated enhanced anti-tumor capacity. In conclusion, ERO1L is overexpressed in CRC, and ERO1L deficiency induces apoptosis and autophagy via ER stress. ERO1L inhibition combined with ER stress-inducing therapies exhibits more effective anti-tumor activity against CRC. ERO1L may serve as a biomarker and therapeutic target for CRC treatment.
Collapse
Affiliation(s)
- Peng Chen
- Department of Integrated Oncology, Center for Integrated Oncology (CIO), University Hospital Bonn, 53127 Bonn, Germany
| | - Yinhao Chen
- Department of Integrated Oncology, Center for Integrated Oncology (CIO), University Hospital Bonn, 53127 Bonn, Germany
| | - Amit Sharma
- Department of Integrated Oncology, Center for Integrated Oncology (CIO), University Hospital Bonn, 53127 Bonn, Germany; Department of Neurosurgery, University Hospital Bonn, 53127 Bonn, Germany
| | | | - Ingo G H Schmidt-Wolf
- Department of Integrated Oncology, Center for Integrated Oncology (CIO), University Hospital Bonn, 53127 Bonn, Germany.
| |
Collapse
|
34
|
Saadh MJ, Ehymayed HM, Alazzawi TS, Fahdil AA, Athab ZH, Yarmukhamedov B, Al-Anbari HHA, Shallal MM, Alsaikhan F, Farhood B. Role of circRNAs in regulating cell death in cancer: a comprehensive review. Cell Biochem Biophys 2025; 83:109-133. [PMID: 39243349 DOI: 10.1007/s12013-024-01492-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/21/2024] [Indexed: 09/09/2024]
Abstract
Despite multiple diagnostic and therapeutic advances, including surgery, radiation therapy, and chemotherapy, cancer preserved its spot as a global health concern. Prompt cancer diagnosis, treatment, and prognosis depend on the discovery of new biomarkers and therapeutic strategies. Circular RNAs (circRNAs) are considered as a stable, conserved, abundant, and varied group of RNA molecules that perform multiple roles such as gene regulation. There is evidence that circRNAs interact with RNA-binding proteins, especially capturing miRNAs. An extensive amount of research has presented the substantial contribution of circRNAs in various types of cancer. To fully understand the linkage between circRNAs and cancer growth as a consequence of various cell death processes, including autophagy, ferroptosis, and apoptosis, more research is necessary. The expression of circRNAs could be controlled to limit the occurrence and growth of cancer, providing a more encouraging method of cancer treatment. Consequently, it is critical to understand how circRNAs affect various forms of cancer cell death and evaluate whether circRNAs could be used as targets to induce tumor death and increase the efficacy of chemotherapy. The current study aims to review and comprehend the effects that circular RNAs exert on cell apoptosis, autophagy, and ferroptosis in cancer to investigate potential cancer treatment targets.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | | | - Tuqa S Alazzawi
- College of dentist, National University of Science and Technology, Dhi Qar, Iraq
| | - Ali A Fahdil
- Medical technical college, Al-Farahidi University, Baghdad, Iraq
| | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - Bekhzod Yarmukhamedov
- Department of Surgical Dentistry and Dental Implantology, Tashkent State Dental Institute, Tashkent, Uzbekistan
- Department of Scientific affairs, Samarkand State Medical University, Samarkand, Uzbekistan
| | | | | | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia.
- School of Pharmacy, Ibn Sina National College for Medical Studies, Jeddah, Saudi Arabia.
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
35
|
Huang WQ, You W, Zhu YQ, Gao F, Wu ZZ, Chen G, Xiao J, Shao Q, Wang LH, Nie X, Zhang Z, Hong CY, You YZ. Autophagosomes coated in situ with nanodots act as personalized cancer vaccines. NATURE NANOTECHNOLOGY 2025; 20:451-462. [PMID: 39753731 DOI: 10.1038/s41565-024-01826-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 10/14/2024] [Indexed: 03/20/2025]
Abstract
Autophagosome cancer vaccines can promote cross-presentation of multiple tumour antigens and induce cross-reactive T cell responses. However, so far, there is no effective method for obtaining a highly immunogenic autophagosomal cancer vaccine because autophagosomes, once formed, quickly fuse with lysosomes and cannot easily escape from cells. Here we report a functional Ti2NX nanodot that caps the autophagosome membrane lipid phosphatidylinositol-4-phosphate, blocking the fusion of autophagosomes with lysosomes and producing stable nanodot-coated autophagosomes in tumours. The formed nanodot-coated autophagosomes can escape from cancer cells to lymph nodes, where they activate tumour-specific T cells. We show that our approach reduces tumour burden and provide long-term immune surveillance protection for cured mice. This work provides a method for the direct formation of personalized autophagosome-based cancer vaccines in vivo, offering a promising strategy for tumour treatment.
Collapse
Affiliation(s)
- Wei-Qiang Huang
- Department of Urology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Hefei National Research Centre for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China
- Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, China
| | - Wei You
- Hefei National Research Centre for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China
- Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, China
| | - Ya-Qi Zhu
- Department of Urology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Hefei National Research Centre for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China
| | - Fan Gao
- Hefei National Research Centre for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China
- Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, China
| | - Zhi-Zhi Wu
- Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, China
| | - Guang Chen
- Hefei National Research Centre for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China.
- Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, China.
| | - Jun Xiao
- Department of Urology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Qi Shao
- Hefei National Research Centre for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China
- Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, China
| | - Long-Hai Wang
- Hefei National Research Centre for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China.
- Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, China.
| | - Xuan Nie
- Hefei National Research Centre for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China
- Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, China
| | - Ze Zhang
- Hefei National Research Centre for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China
- Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, China
| | - Chun-Yan Hong
- Hefei National Research Centre for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China.
- Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, China.
| | - Ye-Zi You
- Department of Urology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Hefei National Research Centre for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China.
- Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
36
|
Morsy MM, Hassan HA, Morsi RM, Nafea OE, Farag AI, Ramadan RS. Alogliptin attenuates testicular damage induced by monosodium glutamate in both juvenile and adult male rats by activating autophagy: ROS dependent AMPK/mTOR. Reprod Toxicol 2025; 132:108826. [PMID: 39725177 DOI: 10.1016/j.reprotox.2024.108826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 12/20/2024] [Accepted: 12/20/2024] [Indexed: 12/28/2024]
Abstract
Monosodium glutamate (MSG) is one of the most commonly used food additives, known for its adverse health effects. Alogliptin (ALO) is a highly selective dipeptidyl peptidase-4 inhibitor, but its role in male reproductive function remains debated. The study was designed to evaluate and compare the potential of ALO in mitigating MSG-induced testicular toxicity in juvenile and adult male rats. Juvenile and adult male rats were treated with either MSG or pretreated with ALO before MSG administration. The rats then received ALO and MSG concurrently for 28 days. Testicular tissues were isolated and subjected to histo-biochemical and molecular assessments. Our results demonstrated that ALO reversed MSG-induced testicular injury, as evidenced by the restoration of reproductive hormone balance (increased serum luteinizing hormone and testosterone concentrations), suppression of oxidative stress injury (decreased testicular malondialdehyde, increased superoxide dismutase activity, and minimal 8-hydroxy-2'-deoxyguanosine immunoreactivity), inflammation (reduced testicular tumor necrosis factor-alpha levels), and fibrosis (decreased testicular collagen fiber deposition). Additionally, ALO impeded apoptosis and activated autophagy by decreasing caspase-3 activity, stimulating the AMPK/mTOR pathway, downregulating Bax and SQSTM-1/p62 expression, upregulating Bcl2 and Beclin 1, promoting testicular proliferation (increased number of proliferating cell nuclear antigen-positive cells in the testis), restoring glycogen content in the testis (mild to moderate periodic acid-Schiff reaction), and preserving testicular architecture. MSG induced more severe adverse testicular effects in juvenile rats, while ALO pretreatment was more protective in adult rats. ALO's anti-inflammatory, antioxidant, antiapoptotic, pro-autophagic, antifibrotic, and proliferative actions in the testis suggest its promising potential for combating male reproductive dysfunction.
Collapse
Affiliation(s)
- Manal Mohammad Morsy
- Human Anatomy and Embryology Department, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Heba A Hassan
- Clinical Pharmacology Department, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt; Pharmacology Department, Faculty of Medicine, Mutah University, Mutah 61710, Jordan
| | - Reham M Morsi
- Biological Application Department, Nuclear Research Center, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Ola Elsayed Nafea
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt.
| | - Azza I Farag
- Human Anatomy and Embryology Department, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt; Department of Physical Therapy, College of Applied Medical Sciences, Qassim University, P.O. Box 6666, Buraydah 51452, Saudi Arabia
| | - Rania Saad Ramadan
- Human Anatomy and Embryology Department, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt; Department of Anatomy, College of Medicine, Al-Baha University, Al-Baha 65525, Saudi Arabia
| |
Collapse
|
37
|
Angel-Lerma LE, Carrillo-Campos J, Siañez-Estrada LI, Siqueiros-Cendón TS, León-Flores DB, Espinoza-Sánchez EA, Arévalo-Gallegos S, Iglesias-Figueroa BF, Rascón-Cruz Q. Molecular Docking of Lactoferrin with Apoptosis-Related Proteins Insights into Its Anticancer Mechanism. Int J Mol Sci 2025; 26:2023. [PMID: 40076649 PMCID: PMC11899785 DOI: 10.3390/ijms26052023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 02/20/2025] [Accepted: 02/22/2025] [Indexed: 03/14/2025] Open
Abstract
Human Lactoferrin (hLf), a multifunctional glycoprotein, has been analyzed through molecular docking to evaluate its role in apoptosis regulation and its potential as an anticancer agent. The docking results highlight XIAP (X-linked Inhibitor of Apoptosis Protein) and Caspase-3 as the most reliable targets, where hLf disrupts XIAP's inhibition of Caspase-3 and Caspase-9, potentially restoring apoptotic signaling; hLf also stabilizes Caspase-3, enhancing its activation in intrinsic and extrinsic pathways. Weaker interactions were observed with Fas, Bcl-2, and Akt. hLf's role in Fas signaling is likely due to expression upregulation rather than direct binding. In contrast, its binding to Bcl-2 may disrupt anti-apoptotic function, and its interaction with Akt suggests interference with pro-survival signaling. These findings suggest that hLf may promote apoptosis by enhancing caspase activation and modulating key apoptotic regulators, supporting its potential use in cancer treatment. However, further experimental validation is needed to confirm these interactions and their therapeutic implications.
Collapse
Affiliation(s)
- Lidia Esmeralda Angel-Lerma
- Facultad de Ciencias Químicas, Universidad Autónoma de Chihuahua, Campus II Circuito Universitario s/n, Chihuahua 31125, Mexico; (L.E.A.-L.); (L.I.S.-E.); (T.S.S.-C.); (D.B.L.-F.); (E.A.E.-S.); (S.A.-G.); (B.F.I.-F.)
| | - Javier Carrillo-Campos
- Facultad de Zootecnia y Ecología, Universidad Autónoma de Chihuahua, Periférico Francisco R. Almada km 1, Chihuahua 31453, Mexico;
| | - Luis Ignacio Siañez-Estrada
- Facultad de Ciencias Químicas, Universidad Autónoma de Chihuahua, Campus II Circuito Universitario s/n, Chihuahua 31125, Mexico; (L.E.A.-L.); (L.I.S.-E.); (T.S.S.-C.); (D.B.L.-F.); (E.A.E.-S.); (S.A.-G.); (B.F.I.-F.)
| | - Tania Samanta Siqueiros-Cendón
- Facultad de Ciencias Químicas, Universidad Autónoma de Chihuahua, Campus II Circuito Universitario s/n, Chihuahua 31125, Mexico; (L.E.A.-L.); (L.I.S.-E.); (T.S.S.-C.); (D.B.L.-F.); (E.A.E.-S.); (S.A.-G.); (B.F.I.-F.)
| | - Dyada Blanca León-Flores
- Facultad de Ciencias Químicas, Universidad Autónoma de Chihuahua, Campus II Circuito Universitario s/n, Chihuahua 31125, Mexico; (L.E.A.-L.); (L.I.S.-E.); (T.S.S.-C.); (D.B.L.-F.); (E.A.E.-S.); (S.A.-G.); (B.F.I.-F.)
| | - Edward Alexander Espinoza-Sánchez
- Facultad de Ciencias Químicas, Universidad Autónoma de Chihuahua, Campus II Circuito Universitario s/n, Chihuahua 31125, Mexico; (L.E.A.-L.); (L.I.S.-E.); (T.S.S.-C.); (D.B.L.-F.); (E.A.E.-S.); (S.A.-G.); (B.F.I.-F.)
| | - Sigifredo Arévalo-Gallegos
- Facultad de Ciencias Químicas, Universidad Autónoma de Chihuahua, Campus II Circuito Universitario s/n, Chihuahua 31125, Mexico; (L.E.A.-L.); (L.I.S.-E.); (T.S.S.-C.); (D.B.L.-F.); (E.A.E.-S.); (S.A.-G.); (B.F.I.-F.)
| | - Blanca Flor Iglesias-Figueroa
- Facultad de Ciencias Químicas, Universidad Autónoma de Chihuahua, Campus II Circuito Universitario s/n, Chihuahua 31125, Mexico; (L.E.A.-L.); (L.I.S.-E.); (T.S.S.-C.); (D.B.L.-F.); (E.A.E.-S.); (S.A.-G.); (B.F.I.-F.)
| | - Quintín Rascón-Cruz
- Facultad de Ciencias Químicas, Universidad Autónoma de Chihuahua, Campus II Circuito Universitario s/n, Chihuahua 31125, Mexico; (L.E.A.-L.); (L.I.S.-E.); (T.S.S.-C.); (D.B.L.-F.); (E.A.E.-S.); (S.A.-G.); (B.F.I.-F.)
| |
Collapse
|
38
|
Li S, Mingoia S, Montégut L, Lambertucci F, Chen H, Dong Y, De Palma FDE, Scuderi SA, Rong Y, Carbonnier V, Martins I, Maiuri MC, Kroemer G. Atlas of expression of acyl CoA binding protein/diazepam binding inhibitor (ACBP/DBI) in human and mouse. Cell Death Dis 2025; 16:134. [PMID: 40011442 PMCID: PMC11865319 DOI: 10.1038/s41419-025-07447-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 01/14/2025] [Accepted: 02/11/2025] [Indexed: 02/28/2025]
Abstract
Acyl CoA binding protein encoded by diazepam binding inhibitor (ACBP/DBI) is a tissue hormone that stimulates lipo-anabolic responses and inhibits autophagy, thus contributing to aging and age-related diseases. Protein expression profiling of ACBP/DBI was performed on mouse tissues to identify organs in which this major tissue hormone is expressed. Transcriptomic and proteomic data bases corroborated a high level of human-mouse interspecies conservation of ACBP/DBI expression in different organs. Single-cell RNA-seq data confirmed that ACBP/DBI was strongly expressed by parenchymatous cells from specific human and mouse organs (e.g., kidney, large intestine, liver, lung) as well as by myeloid or glial cells from other organs (e.g., adipose tissue, brain, eye) following a pattern that was conserved among the two species. We identified a panel of 44 mRNAs that are strongly co-expressed with ACBP/DBI mRNA in normal and malignant human and normal mouse tissues. Of note, 22 (50%) of these co-expressed mRNAs encode proteins localized at mitochondria, and mRNAs with metabolism-related functions are strongly overrepresented (66%). Systematic data mining was performed to identify transcription factors that regulate ACBP/DBI expression in human and mouse. Several transcription factors, including growth response 1 (EGR1), E2F Transcription Factor 1 (E2F1, which interacts with retinoblastoma, RB) and transformation-related protein 53 (TRP53, best known as p53), which are endowed with oncosuppressive effects, consistently repress ACBP/DBI expression as well as its co-expressed mRNAs across multiple datasets, suggesting a mechanistic basis for a coregulation network. Furthermore, we identified multiple transcription factors that transactivate ACBP/DBI gene expression together with its coregulation network. Altogether, this study indicates the existence of conserved mechanisms determining the expression of ACBP/DBI in specific cell types of the mammalian organism.
Collapse
Affiliation(s)
- Sijing Li
- Centre de Recherche des Cordeliers, Équipe Labellisée par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Silvia Mingoia
- Centre de Recherche des Cordeliers, Équipe Labellisée par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Department of Pharmacological Sciences, University of Piemonte Orientale, Novara, Italy
| | - Léa Montégut
- Centre de Recherche des Cordeliers, Équipe Labellisée par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Flavia Lambertucci
- Centre de Recherche des Cordeliers, Équipe Labellisée par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Hui Chen
- Centre de Recherche des Cordeliers, Équipe Labellisée par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Yanbing Dong
- Centre de Recherche des Cordeliers, Équipe Labellisée par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Faculté de Médecine, Université de Paris Saclay, Kremlin Bicêtre, Paris, France
| | - Fatima Domenica Elisa De Palma
- Centre de Recherche des Cordeliers, Équipe Labellisée par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Department of Molecular Medicine and Medical Biotechnologies, University of Napoli Federico II, Napoli, Italy
| | - Sarah Adriana Scuderi
- Centre de Recherche des Cordeliers, Équipe Labellisée par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Yan Rong
- Centre de Recherche des Cordeliers, Équipe Labellisée par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Faculté de Médecine, Université de Paris Saclay, Kremlin Bicêtre, Paris, France
| | - Vincent Carbonnier
- Centre de Recherche des Cordeliers, Équipe Labellisée par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Isabelle Martins
- Centre de Recherche des Cordeliers, Équipe Labellisée par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Maria Chiara Maiuri
- Centre de Recherche des Cordeliers, Équipe Labellisée par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.
- Department of Molecular Medicine and Medical Biotechnologies, University of Napoli Federico II, Napoli, Italy.
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Équipe Labellisée par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| |
Collapse
|
39
|
Richter S, Lönnecke P, Bovan D, Andrian N, Stoean B, Lehene M, Silaghi-Dumitrescu R, Gaina L, Mijatović S, Maksimović-Ivanić D, Kaluđerović GN, Hey-Hawkins E. Platinum(II/IV) complexes with N-substituted carboxylate ethylenediamine/propylenediamine ligands: preparation, characterization and in vitro activity. Dalton Trans 2025; 54:3597-3609. [PMID: 39775617 DOI: 10.1039/d4dt03041a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
The synthesis and characterization of novel platinum(II) and platinum(IV) complexes derived from unsymmetrical ethylene or propylenediamine derivatives are presented. IR spectroscopy and ESI mass spectrometry techniques were employed to characterize the complexes, revealing distinctive absorption bands and isotope patterns. Furthermore, the complexes were characterized by 1H and 13C NMR spectroscopy. Single-crystal X-ray structural analysis elucidated the coordination geometry and intermolecular interactions of complexes 3, 4 and 6. Cytotoxicity evaluation of the complexes on various cell lines highlighted complex 3 as the most active, realizing its tumoricidal activity through induction of apoptosis and increased total caspase activity in MCF-7 cells. Since its application is followed by cytoprotective autophagy, the effectiveness can be additionally empowered by concomitant inhibition of this process. Furthermore, the PtIV compound 3 induces oxidative stress in hemoglobin, and is reducible by glutathione, suggesting its potential as a carrier for the active PtII precursor 2a to cancer cells without increasing cytotoxicity. Cyclic voltammetry corroborates the ability of complex 3 to undergo reduction under physiological conditions.
Collapse
Affiliation(s)
- Stefan Richter
- Institute of Inorganic Chemistry, Faculty of Chemistry and Mineralogy, Leipzig University, Johannisallee 29, 04103 Leipzig, Germany
| | - Peter Lönnecke
- Institute of Inorganic Chemistry, Faculty of Chemistry and Mineralogy, Leipzig University, Johannisallee 29, 04103 Leipzig, Germany
| | - Dijana Bovan
- Department of Immunology, Institute for Biological Research "Siniša Stanković" National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11060 Belgrade, Serbia
| | - Nicoleta Andrian
- Department of Chemistry, Babeş-Bolyai University, Str. Arany Janos Nr. 11, RO-400028 Cluj-Napoca, Romania
| | - Bianca Stoean
- Department of Chemistry, Babeş-Bolyai University, Str. Arany Janos Nr. 11, RO-400028 Cluj-Napoca, Romania
| | - Maria Lehene
- Department of Chemistry, Babeş-Bolyai University, Str. Arany Janos Nr. 11, RO-400028 Cluj-Napoca, Romania
| | - Radu Silaghi-Dumitrescu
- Department of Chemistry, Babeş-Bolyai University, Str. Arany Janos Nr. 11, RO-400028 Cluj-Napoca, Romania
| | - Luiza Gaina
- Department of Chemistry, Babeş-Bolyai University, Str. Arany Janos Nr. 11, RO-400028 Cluj-Napoca, Romania
| | - Sanja Mijatović
- Department of Immunology, Institute for Biological Research "Siniša Stanković" National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11060 Belgrade, Serbia
| | - Danijela Maksimović-Ivanić
- Department of Immunology, Institute for Biological Research "Siniša Stanković" National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11060 Belgrade, Serbia
| | - Goran N Kaluđerović
- Department of Engineering and Natural Sciences, University of Applied Sciences Merseburg, Eberhard Leibnitz-Str. 2, 06217 Merseburg, Germany
| | - Evamarie Hey-Hawkins
- Department of Chemistry, Babeş-Bolyai University, Str. Arany Janos Nr. 11, RO-400028 Cluj-Napoca, Romania
- Universität Leipzig, Faculty of Chemistry and Mineralogy, Institute of Bioanalytical Chemistry, BBZ, Deutscher Platz 5, 04103 Leipzig, Germany.
| |
Collapse
|
40
|
Cabrera-Serrano AJ, Sánchez-Maldonado JM, González-Olmedo C, Carretero-Fernández M, Díaz-Beltrán L, Gutiérrez-Bautista JF, García-Verdejo FJ, Gálvez-Montosa F, López-López JA, García-Martín P, Pérez EM, Sánchez-Rovira P, Reyes-Zurita FJ, Sainz J. Crosstalk Between Autophagy and Oxidative Stress in Hematological Malignancies: Mechanisms, Implications, and Therapeutic Potential. Antioxidants (Basel) 2025; 14:264. [PMID: 40227235 PMCID: PMC11939785 DOI: 10.3390/antiox14030264] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/19/2025] [Accepted: 02/19/2025] [Indexed: 04/15/2025] Open
Abstract
Autophagy is a fundamental cellular process that maintains homeostasis by degrading damaged components and regulating stress responses. It plays a crucial role in cancer biology, including tumor progression, metastasis, and therapeutic resistance. Oxidative stress, similarly, is key to maintaining cellular balance by regulating oxidants and antioxidants, with its disruption leading to molecular damage. The interplay between autophagy and oxidative stress is particularly significant, as reactive oxygen species (ROS) act as both inducers and by-products of autophagy. While autophagy can function as a tumor suppressor in early cancer stages, it often shifts to a pro-tumorigenic role in advanced disease, aiding cancer cell survival under adverse conditions such as hypoxia and nutrient deprivation. This dual role is mediated by several signaling pathways, including PI3K/AKT/mTOR, AMPK, and HIF-1α, which coordinate the balance between autophagic activity and ROS production. In this review, we explore the mechanisms by which autophagy and oxidative stress interact across different hematological malignancies. We discuss how oxidative stress triggers autophagy, creating a feedback loop that promotes tumor survival, and how autophagic dysregulation leads to increased ROS accumulation, exacerbating tumorigenesis. We also examine the therapeutic implications of targeting the autophagy-oxidative stress axis in cancer. Current strategies involve modulating autophagy through specific inhibitors, enhancing ROS levels with pro-oxidant compounds, and combining these approaches with conventional therapies to overcome drug resistance. Understanding the complex relationship between autophagy and oxidative stress provides critical insights into novel therapeutic strategies aimed at improving cancer treatment outcomes.
Collapse
Affiliation(s)
- Antonio José Cabrera-Serrano
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Instituto de Investigación Biosanitaria IBs.Granada, 18012 Granada, Spain;
| | - José Manuel Sánchez-Maldonado
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Instituto de Investigación Biosanitaria IBs.Granada, 18012 Granada, Spain;
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, 18012 Granada, Spain
| | - Carmen González-Olmedo
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Medical Oncology Unit, University Hospital of Jaén, 23007 Jaén, Spain
| | - María Carretero-Fernández
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Instituto de Investigación Biosanitaria IBs.Granada, 18012 Granada, Spain;
| | - Leticia Díaz-Beltrán
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Medical Oncology Unit, University Hospital of Jaén, 23007 Jaén, Spain
| | - Juan Francisco Gutiérrez-Bautista
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Instituto de Investigación Biosanitaria IBs.Granada, 18012 Granada, Spain;
- Servicio de Análisis Clínicos e Inmunología, University Hospital Virgen de las Nieves, 18014 Granada, Spain
- Department of Biochemistry, Molecular Biology and Immunology III, University of Granada, 18016 Granada, Spain
| | - Francisco José García-Verdejo
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Medical Oncology Unit, University Hospital of Jaén, 23007 Jaén, Spain
| | - Fernando Gálvez-Montosa
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Medical Oncology Unit, University Hospital of Jaén, 23007 Jaén, Spain
| | - José Antonio López-López
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Medical Oncology Unit, University Hospital of Jaén, 23007 Jaén, Spain
| | - Paloma García-Martín
- Instituto de Investigación Biosanitaria IBs.Granada, 18012 Granada, Spain;
- Campus de la Salud Hospital, PTS, 18016 Granada, Spain
| | - Eva María Pérez
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Instituto de Investigación Biosanitaria IBs.Granada, 18012 Granada, Spain;
- Campus de la Salud Hospital, PTS, 18016 Granada, Spain
| | - Pedro Sánchez-Rovira
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Medical Oncology Unit, University Hospital of Jaén, 23007 Jaén, Spain
| | - Fernando Jesús Reyes-Zurita
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, 18012 Granada, Spain
| | - Juan Sainz
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Instituto de Investigación Biosanitaria IBs.Granada, 18012 Granada, Spain;
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, 18012 Granada, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
| |
Collapse
|
41
|
Gao K, Zhao Y, Si M, Zhang B, Wang Z, Chen H, Lin P, Wang A, Jin Y. ERS regulates endometrial epithelial cell autophagy through XBP1s-mediated activation of the PI3K/AKT pathway. Sci Rep 2025; 15:5943. [PMID: 39966508 PMCID: PMC11836410 DOI: 10.1038/s41598-024-84461-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 12/23/2024] [Indexed: 02/20/2025] Open
Abstract
Autophagy is a fundamental cellular activity involved in the renewal of cellular components, occurring primarily in cells subjected to physiological remodeling or pathological stimuli. The occurrence of autophagy is closely related to the endoplasmic reticulum (ER), and ER stress (ERS) occurs when ER homeostasis is disrupted. The current study aimed to analyze the molecular mechanisms underlying the effects of ERS on autophagy in goat endometrial epithelial cells (gEECs). We found that rapamycin (an autophagy inducer) induced autophagy and ERS in a time-dependent manner in gEECs which was accompanied by significantly increased expression of the autophagy-related genes ATG5, the LC3II/LC3I and ERS-related genes GRP78, IRE1, ATF6, and XBP1s. PI3K and AKT protein phosphorylation was significantly reduced during gEECs autophagy. Interestingly, TG (ERS activator) significantly inhibited the expression of ATG5 and the LC3II/LC3I and significantly promoted expression of SQSTM1, whereas the ERS inhibitor 4-PBA showed the opposite results. Surprisingly, XBP1s knockdown inhibited the effects of TG. Furthermore, XBP1s overexpression significantly inhibited autophagy whereas XBP1s knockdown increased ATG5 expression and the LC3II/LC3I and decreased SQSTM1 expression in gEECs. Specifically, XBP1s overexpression significantly promoted PI3K and AKT protein phosphorylation while treatment with LY294002 (PI3K/AKT pathway inhibitor) significantly reversed the effect. Similarly, PI3K/AKT pathway activation was significantly inhibited following XBP1s knockdown whereas treatment with SC79 (PI3K/AKT pathway activator) showed the opposite results. Taken together, our data suggest that interactions between ERS and autophagy exist in gEECs. XBP1s, the key effector of ERS, inhibits autophagy in gEECs by promoting the PI3K/AKT pathway in gEECs. These results may contribute to the treatment and prevention of uterine diseases.
Collapse
Affiliation(s)
- Kangkang Gao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Yiteng Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Mengqi Si
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Beibei Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Zongjie Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Huatao Chen
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Pengfei Lin
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Aihua Wang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Yaping Jin
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China.
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| |
Collapse
|
42
|
Lee GH, Lee SH, Li XH, Lu QY, Zhan CL, Kim JD, Sim JM, Song HJ, Sun MH, Cui XS. ERK5 is essential for early porcine embryonic development by maintaining Endoplasmic Reticulum homeostasis. Gene 2025; 936:149104. [PMID: 39557370 DOI: 10.1016/j.gene.2024.149104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 10/13/2024] [Accepted: 11/15/2024] [Indexed: 11/20/2024]
Abstract
Extracellular signal-regulated kinase 5 (ERK5), a mitogen-activated protein kinase (MAPK) family member, plays an important role in various biological processes, such as proliferation, apoptosis, differentiation, survival, and cell regulation. However, studies on the effects of ERK5 on porcine preimplantation embryos are limited. In this study, to determine the function of ERK5 during porcine embryo development, ERK5 function was inhibited by adding the ERK5 inhibitor JWG-071. The ERK5 mRNA and protein expression levels tended to decrease from the 4-cell stage compared to the 1-cell and 2-cell stages, suggesting that ERK5 is the maternal gene. During embryonic development in pigs, adding 5 μM of JWG-071 significantly reduced the phosphorylation of ERK5 and the blastocyst development rate (control: 53.44 ± 8.38 %; treatment: 26.65 ± 3.40 %). Additionally, ERK5 inhibition increased the expression of UPR-related proteins, glucose-regulated protein (GRP78), and C/EBP homologous protein (CHOP) by inducing ER stress. Compared to the control group, the expression of the autophagy-related proteins LC3 and ATG7 was significantly increased in the ERK5 inhibition group, indicating that the inhibition of ERK5 induced autophagy. In addition, ERK5 inhibition increased the expression of BAX, a pro-apoptotic gene, resulting in apoptosis. In conclusion, the results show that ERK5 inhibition during porcine embryonic development induces autophagy and apoptosis by increasing ER stress, resulting in a negative effect on embryonic development in pigs.
Collapse
Affiliation(s)
- Gyu-Hyun Lee
- Department of Animal Science, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Song-Hee Lee
- Department of Animal Science, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Xiao-Han Li
- Department of Animal Science, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Qin-Yue Lu
- Department of Animal Science, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Cheng-Lin Zhan
- Department of Animal Science, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Ji-Dam Kim
- Department of Animal Science, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Jae-Min Sim
- Department of Animal Science, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Hyeon-Ji Song
- Department of Animal Science, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Ming-Hong Sun
- Department of Animal Science, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea; College of Animal Science and Technology, Southwest University, Chongqing 402460, China
| | - Xiang-Shun Cui
- Department of Animal Science, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea.
| |
Collapse
|
43
|
Yan Z, Zheng Z, Cao L, Zhu Z, Zhou C, Sun Q, Tang B, Zhao G. Altered gut microbiome and serum metabolome profiles associated with essential tremor. Metab Brain Dis 2025; 40:118. [PMID: 39903340 DOI: 10.1007/s11011-025-01549-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 01/30/2025] [Indexed: 02/06/2025]
Abstract
The genetic predisposition and environmental factors both trigger the complex neurological dyskinesia of essential tremor (ET). Gut dysbiosis may facilitate the occurrence and development of neurological diseases. Therefore, it is worth exploring the inner connections between gut microbiota and ET. First, the gut microbiota of 19 ET patients and 21 healthy controls (HCs) were analysed with metagenomics approach. Second, the potential linkages between gut microbiome and serum metabolome profiles were explored by integrative analysis. The gut microbiota disorders were present in ET patients. The LEfSe method showed a significant decrease in Bacteroides. The functional analysis revealed that there were differences in gut microbial apoptosis, retinol metabolism, and steroid hormone biosynthesis pathways. The levels of various lipids and lipid-like molecules alter in serum of ET patients, which correlated with altered gut microbial abundance, indicating the alterations in lipid metabolism involved in apoptosis pathway in ET. All of these data point to the gut dysbiosis in ET, and some changed gut microbial species were linked to abnormalities in blood lipid metabolism, which open up new avenues for investigation into the pathophysiology of ET.
Collapse
Affiliation(s)
- Zhenzhen Yan
- Department of Neurology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, Zhejiang, P.R. China
| | - Zhilin Zheng
- Department of Neurology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, Zhejiang, P.R. China
| | - Lanxiao Cao
- Department of Neurology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, Zhejiang, P.R. China
| | - Zeyu Zhu
- Department of Neurology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, Zhejiang, P.R. China
| | - Chen Zhou
- Department of Neurology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, Zhejiang, P.R. China
| | - Qiying Sun
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, P.R. China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, P.R. China
| | - Guohua Zhao
- Department of Neurology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, Zhejiang, P.R. China.
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China.
| |
Collapse
|
44
|
Qi G, Ma H, Teng K, Gai P, Gong Y, Chen J, Luo X, Kong B. SHCBP1 promotes cisplatin resistance of ovarian cancer through AKT/mTOR/Autophagy pathway. Apoptosis 2025; 30:83-98. [PMID: 39397124 DOI: 10.1007/s10495-024-02027-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2024] [Indexed: 10/15/2024]
Abstract
Ovarian cancer caused the highest cancer-related mortality among female reproductive system malignancies. Platinum-based chemotherapy is still the footstone of the chemotherapy for ovarian cancer. However, the molecular mechanisms underlying cisplatin insensitivity and resistance remain unclear. SHC SH2 domain-binding protein 1 (SHCBP1) plays critical roles in the progression and drug resistance of different types of cancer. However, the biological function of SHCBP1 in ovarian cancer progression and cisplatin resistance remains obscure. In this study, we found that SHCBP1 was upregulated in ovarian cancer and the upregulated SHCBP1 has growth-promoting effect on ovarian cancer cells. Furthermore, SHCBP1 silencing sensitize ovarian cancer cells to cisplatin (hereafter referred to as CDDP). Mechanism analysis revealed that SHCBP1 activated the Akt/mTOR pathway and further inhibited autophagy in ovarian cancer cells. Meanwhile, autophagy inhibitors combined with SHCBP1 knockdown enhances CDDP sensitivity. In addition, knockdown of SHCBP1 restricted the proliferation of tumors and increased the cisplatin sensitivity in vivo. These findings suggested that upregulated SHCBP1 promoted the proliferation and CDDP resistance of ovarian cancer. The combination of SHCBP1 inhibition and cisplatin treatment might lead to substantial progress in ovarian cancer targeted therapy.
Collapse
Affiliation(s)
- Gonghua Qi
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, 107 Wenhua Xi Road, Jinan, 250012, China
- Gynecologic Oncology Key Laboratory of Shandong Province, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Hanlin Ma
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, 107 Wenhua Xi Road, Jinan, 250012, China
- Gynecologic Oncology Key Laboratory of Shandong Province, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Kai Teng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, 250117, China
| | - Panpan Gai
- 71217 of the Chinese People's Liberation Army, Laiyang, 265200, China
| | - Yanmin Gong
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, 107 Wenhua Xi Road, Jinan, 250012, China
| | - Jingying Chen
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, 107 Wenhua Xi Road, Jinan, 250012, China
- Gynecologic Oncology Key Laboratory of Shandong Province, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Xia Luo
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, 107 Wenhua Xi Road, Jinan, 250012, China.
| | - Beihua Kong
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, 107 Wenhua Xi Road, Jinan, 250012, China.
- Gynecologic Oncology Key Laboratory of Shandong Province, Qilu Hospital, Shandong University, Jinan, 250012, China.
| |
Collapse
|
45
|
Tagne AM, Fotio Y, Lee HL, Jung KM, Katz J, Ahmed F, Le J, Bazinet R, Jang C, Piomelli D. Metabolic reprogramming in the spinal cord drives the transition to pain chronicity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.30.635746. [PMID: 39975205 PMCID: PMC11838349 DOI: 10.1101/2025.01.30.635746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Acute injuries can progress into painful states that endure long after healing. The mechanism underlying this transition remains unclear, but metabolic adaptations to the bioenergy demands imposed by injury are plausible contributors. Here we show that peripheral injury activates AKT/mTORC1 in afferent segments of the mouse spinal cord, redirecting local core metabolism toward biomass production while simultaneously suppressing autophagy-mediated biomass reclamation. This metabolic shift supports neuroplasticity, but creates a resource bottleneck that depletes critical spinal cord nutrients. Preventing this depletion with a modified diet normalizes biomass generation and autophagy and halts the transition to chronic pain. This effect, observed across multiple pain models, requires activation of the nutrient sensors, sirtuin-1 and AMPK, as well as restoration of autophagy. The findings identify metabolic reprogramming as a key driver of the progression to pain chronicity and point to nutritional and pharmacological interventions that could prevent this progression after surgery or other physical traumas.
Collapse
Affiliation(s)
- Alex Mabou Tagne
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
| | - Yannick Fotio
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
| | - Hye-Lim Lee
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
| | - Kwang-Mook Jung
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
| | - Jean Katz
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
| | - Faizy Ahmed
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
| | - Johnny Le
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Richard Bazinet
- Department of Nutritional Sciences, University of Toronto, ON, Canada
| | - Cholsoon Jang
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Daniele Piomelli
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
- Department of Pharmaceutical Sciences, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
46
|
Samanta A, Biswas S, Ghosh S, Banerjee S, Dam B, Maitra S. Maternal exposure to chronic, low-dose nonylphenol in zebrafish: Disruption of ovarian health, reproductive function, and embryogenesis. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2025; 375:124169. [PMID: 39842349 DOI: 10.1016/j.jenvman.2025.124169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 01/07/2025] [Accepted: 01/15/2025] [Indexed: 01/24/2025]
Abstract
Nonylphenol (NP), a non-ionic surfactant and potent endocrine disruptor, is known for its environmental persistence, biotic accumulation potential and toxicity. Nonetheless, mechanisms underlying NP modulation of female fertility with potential impact on embryogenesis in the unexposed offspring remain elusive. This study investigates the effects and toxic mechanisms of maternal exposure to NP at varying concentrations (50 and 100 μg/L) on zebrafish (Danio rerio), specifically focusing on ovarian health, reproductive parameters, and early developmental potential in the F1 generation. Our findings indicate a higher accumulation of NP in the ovaries compared to muscle tissue. Further, chronic (28 days) NP exposure promotes ovarian reactive oxygen species (ROS) accumulation, activates the MAPK (JNK, p38 MAPK, ERK1/2) pathways, AP-1 induction, and elevated expression of pro-inflammatory cytokines (Tnf-α, Il-1β, Il-6) triggering inflammation. Besides, heightened follicular atresia in NP-treated ovaries relates to increased Bax/Bcl2 ratio, cleaved caspase 3 and Parp1 activation prompting apoptosis. While it showed higher affinity to zebrafish ERα (in silico analysis), NP exposure in vivo promotes a robust increase in ovarian ERα but abrogated ERβ expression and a significant alteration in fshr and lhcgr transcripts. While attenuated StAR and P450 aromatase expression at both mRNA and protein levels and reduced igf3 expression reveal impaired ovarian microenvironment, NP-induced dysregulated NO/NOS/cyclooxygenase signaling and attenuation of hCG-induced p34cdc2 activation and oocyte maturation correspond well with decreased fecundity and fertilization efficiency. Intriguingly, maternal exposure to NP resulted in delayed embryogenesis, developmental aberrations, and reduced hatching rates in the unexposed offspring, risking F1 generation. Collectively, this study provides mechanistic insights into the detrimental influence of maternal exposure to NP on ovarian dysfunction, reproductive insufficiency and embryotoxicity.
Collapse
Affiliation(s)
- Anwesha Samanta
- Molecular and Cellular Endocrinology Laboratory, Department of Zoology, Visva-Bharati University, Santiniketan, 731235, India
| | - Subhasri Biswas
- Molecular and Cellular Endocrinology Laboratory, Department of Zoology, Visva-Bharati University, Santiniketan, 731235, India
| | - Sandip Ghosh
- Microbiology Laboratory, Department of Botany, Visva-Bharati University, Santiniketan, 731235, India
| | - Sambuddha Banerjee
- Molecular and Cellular Endocrinology Laboratory, Department of Zoology, Visva-Bharati University, Santiniketan, 731235, India
| | - Bomba Dam
- Microbiology Laboratory, Department of Botany, Visva-Bharati University, Santiniketan, 731235, India
| | - Sudipta Maitra
- Molecular and Cellular Endocrinology Laboratory, Department of Zoology, Visva-Bharati University, Santiniketan, 731235, India.
| |
Collapse
|
47
|
Khaliulin I, Hamoudi W, Amal H. The multifaceted role of mitochondria in autism spectrum disorder. Mol Psychiatry 2025; 30:629-650. [PMID: 39223276 PMCID: PMC11753362 DOI: 10.1038/s41380-024-02725-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
Normal brain functioning relies on high aerobic energy production provided by mitochondria. Failure to supply a sufficient amount of energy, seen in different brain disorders, including autism spectrum disorder (ASD), may have a significant negative impact on brain development and support of different brain functions. Mitochondrial dysfunction, manifested in the abnormal activities of the electron transport chain and impaired energy metabolism, greatly contributes to ASD. The aberrant functioning of this organelle is of such high importance that ASD has been proposed as a mitochondrial disease. It should be noted that aerobic energy production is not the only function of the mitochondria. In particular, these organelles are involved in the regulation of Ca2+ homeostasis, different mechanisms of programmed cell death, autophagy, and reactive oxygen and nitrogen species (ROS and RNS) production. Several syndromes originated from mitochondria-related mutations display ASD phenotype. Abnormalities in Ca2+ handling and ATP production in the brain mitochondria affect synaptic transmission, plasticity, and synaptic development, contributing to ASD. ROS and Ca2+ regulate the activity of the mitochondrial permeability transition pore (mPTP). The prolonged opening of this pore affects the redox state of the mitochondria, impairs oxidative phosphorylation, and activates apoptosis, ultimately leading to cell death. A dysregulation between the enhanced mitochondria-related processes of apoptosis and the inhibited autophagy leads to the accumulation of toxic products in the brains of individuals with ASD. Although many mitochondria-related mechanisms still have to be investigated, and whether they are the cause or consequence of this disorder is still unknown, the accumulating data show that the breakdown of any of the mitochondrial functions may contribute to abnormal brain development leading to ASD. In this review, we discuss the multifaceted role of mitochondria in ASD from the various aspects of neuroscience.
Collapse
Affiliation(s)
- Igor Khaliulin
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Wajeha Hamoudi
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Haitham Amal
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
48
|
Mićević M, Čalija S, Korićanac L, Žakula J, Vilotić A, Radović M, Golić I, Korać A, Nacka-Aleksić M, Stojadinović B, Dohčević-Mitrović Z. Probing the effects of dextran-coated CeO 2 nanoparticles on lung fibroblasts using multivariate single-cell Raman spectroscopy. Nanotoxicology 2025; 19:100-118. [PMID: 39840838 DOI: 10.1080/17435390.2025.2453576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 12/17/2024] [Accepted: 01/02/2025] [Indexed: 01/23/2025]
Abstract
In this study, we investigated the cytotoxic effect of highly soluble dextran-coated CeO2 nanoparticles on human fetal lung fibroblasts MRC-5. We examined individual nanoparticle-treated cells by Raman spectroscopy and analyzed Raman spectra using non-negative principal component analysis and k-means clustering. In this way, we determined dose-dependent differences between treated cells, which were reflected through the intensity change of lipid, phospholipid and RNA-related Raman modes. Performing standard biological tests for cell growth, viability and induction of apoptosis in parallel, these changes were correlated with nanoparticle-induced apoptotic processes. The cells with specific spectral characteristics, referring to non-apoptotic, but possibly autophagic cell death modality, were also detected. Additionally, Raman imaging combined with principal component and vertex component analysis was used to map the spatial distribution of biological molecules in treated and untreated cells. This work provided the description of different resulting states of the treated cells depending on the dextran-coated CeO2 nanoparticles dose, which can be later used in the design of the nanoparticles for industrial or medical applications. The wide content of information resulting from single-cell Raman spectroscopy has the potential to detect biochemical changes caused by nanoparticles that would otherwise require a series of expensive and time-consuming standard biological techniques.
Collapse
Affiliation(s)
- Mirjana Mićević
- Institute of Physics Belgrade, University of Belgrade, Belgrade, Serbia
| | - Sonja Čalija
- Institute of Physics Belgrade, University of Belgrade, Belgrade, Serbia
| | - Lela Korićanac
- Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Jelena Žakula
- Vinča Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Aleksandra Vilotić
- Institute for the Application of Nuclear Energy, Department for Biology of Reproduction, University of Belgrade, Belgrade, Serbia
| | - Marko Radović
- BioSense Institute, University of Novi Sad, Novi Sad, Serbia
| | - Igor Golić
- Center for Electron Microscopy, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Aleksandra Korać
- Center for Electron Microscopy, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Mirjana Nacka-Aleksić
- Institute for the Application of Nuclear Energy, Department for Biology of Reproduction, University of Belgrade, Belgrade, Serbia
| | | | | |
Collapse
|
49
|
Zhang W, Wu H, Liao Y, Zhu C, Zou Z. Caspase family in autoimmune diseases. Autoimmun Rev 2025; 24:103714. [PMID: 39638102 DOI: 10.1016/j.autrev.2024.103714] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/28/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024]
Abstract
Programmed cell death (PCD) plays a crucial role in maintaining tissue homeostasis, with its primary forms including apoptosis, pyroptosis, and necroptosis. The caspase family is central to these processes, and its complex functions across different cell death pathways and other non-cell death roles have been closely linked to the pathogenesis of autoimmune diseases. This article provides a comprehensive review of the role of the caspase family in autoimmune diseases such as rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), type 1 diabetes (T1D), and multiple sclerosis (MS). It particularly emphasizes the intricate functions of caspases within various cell death pathways and their potential as therapeutic targets, thereby offering innovative insights and a thorough discussion in this field. In terms of therapy, strategies targeting caspases hold significant promise. We emphasize the importance of a holistic understanding of caspases in the overall concept of cell death, exploring their unique functions and interrelationships across multiple cell death pathways, including apoptosis, pyroptosis, necroptosis, and PANoptosis. This approach transcends the limitations of previous studies that focused on singular cell death pathways. Additionally, caspases play a key role in non-cell death functions, such as immune cell activation, cytokine processing, inflammation regulation, and tissue repair, thereby opening new avenues for the treatment of autoimmune diseases. Regulating caspase activity holds the potential to restore immune balance in autoimmune diseases. Potential therapeutic approaches include small molecule inhibitors (both reversible and irreversible), biological agents (such as monoclonal antibodies), and gene therapies. However, achieving specific modulation of caspases to avoid interference with normal physiological functions remains a major challenge. Future research must delve deeper into the regulatory mechanisms of caspases and their associated complexes linked to PANoptosis to facilitate precision medicine. In summary, this article offers a comprehensive and in-depth analysis, providing a novel perspective on the complex roles of caspases in autoimmune diseases, with the potential to catalyze breakthroughs in understanding disease mechanisms and developing therapeutic strategies.
Collapse
Affiliation(s)
- Wangzheqi Zhang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China; School of Anesthesiology, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Huang Wu
- Basic Medical University, Naval Medical University, Shanghai 200433, China
| | - Yan Liao
- School of Anesthesiology, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Chenglong Zhu
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China; School of Anesthesiology, Naval Medical University, 168 Changhai Road, Shanghai 200433, China.
| | - Zui Zou
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China; School of Anesthesiology, Naval Medical University, 168 Changhai Road, Shanghai 200433, China.
| |
Collapse
|
50
|
Chen H, Liu J, Cao Z, Li J, Zhang H, Yang Q, Cheng J, Shen Y, He K. Enhancing hepatocellular carcinoma therapy with DOX-loaded SiO 2 nanoparticles via mTOR-TFEB pathway autophagic flux inhibition. J Nanobiotechnology 2025; 23:27. [PMID: 39828690 PMCID: PMC11743218 DOI: 10.1186/s12951-025-03107-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 01/11/2025] [Indexed: 01/22/2025] Open
Abstract
Chemotherapeutic drugs often fail to provide long-term efficacy due to their lack of specificity and high toxicity. To enhance the biosafety and reduce the side effects of these drugs, various nanocarrier delivery systems have been developed. In this study, we loaded the anticancer drug doxorubicin (DOX) and an MRI contrast agent into silica nanoparticles, coating them with pH-responsive and tumor cell-targeting polymers. These polymers enable the carrier to achieve targeted delivery and controlled drug release in acidic environments. This integrated diagnostic and therapeutic strategy successfully achieved both the diagnosis and treatment of liver cancer. Additionally, we demonstrated that the nanocarrier inhibits autophagic flux in liver cancer cells by targeting the autophagy-lysosome pathway and regulating the nuclear translocation of TFEB, thereby promoting tumor cell death. This novel diagnostic-integrated nanocarrier is expected to be a promising tool for targeted liver cancer treatment.
Collapse
Affiliation(s)
- Huanyu Chen
- Imaging Center, The Third Affiliated Hospital of Anhui Medical University, Hefei, 230061, China
- The Fifth Clinical Medical School of Anhui Medical University, Hefei, 230032, China
| | - Jun Liu
- School of Basic Medical Sciences and Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China
| | - Zhichao Cao
- Imaging Center, The Third Affiliated Hospital of Anhui Medical University, Hefei, 230061, China
- Wannan Medical College, Wuhu, 241002, China
| | - Jiajia Li
- Central Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Hong Zhang
- Imaging Center, The Third Affiliated Hospital of Anhui Medical University, Hefei, 230061, China
- Wannan Medical College, Wuhu, 241002, China
| | - Qianqian Yang
- Imaging Center, The Third Affiliated Hospital of Anhui Medical University, Hefei, 230061, China
- The Fifth Clinical Medical School of Anhui Medical University, Hefei, 230032, China
| | - Jian Cheng
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, 215123, China.
| | - Yuxian Shen
- School of Basic Medical Sciences and Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China.
| | - Kewu He
- Imaging Center, The Third Affiliated Hospital of Anhui Medical University, Hefei, 230061, China.
- The Fifth Clinical Medical School of Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|