1
|
Huang X, Xie M, Wang Y, Lu X, Mei F, Zhang K, Yang X, Chen G, Yin Y, Feng G, Song W, Dong N, Deng X, Wang S, Chen L. Porphyromonas gingivalis aggravates atherosclerotic plaque instability by promoting lipid-laden macrophage necroptosis. Signal Transduct Target Ther 2025; 10:171. [PMID: 40404630 PMCID: PMC12098900 DOI: 10.1038/s41392-025-02251-6] [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: 02/11/2024] [Revised: 03/04/2025] [Accepted: 04/28/2025] [Indexed: 05/24/2025] Open
Abstract
At advanced phases of atherosclerosis, the rupture and thrombogenesis of vulnerable plaques emerge as primary triggers for acute cardiovascular events and fatalities. Pathogenic infection such as periodontitis-associated Porphyromonas gingivalis (Pg) has been suspected of increasing the risks of atherosclerotic cardiovascular disease, but its relationship with atherosclerotic plaque destabilization remains elusive. Here we demonstrated that the level of Pg-positive clusters positively correlated with the ratio of necrotic core area to total atherosclerotic plaque area in human clinical samples, which indicates plaque instability. In rabbits and Apoe-/- mice, Pg promoted atherosclerotic plaque necrosis and aggravated plaque instability by triggering oxidative stress, which led to macrophage necroptosis. This process was accompanied by the decreased protein level of forkhead box O3 (FOXO3) in macrophages. The mechanistic dissection showed that Pg lipopolysaccharide (LPS) evoked macrophage oxidative stress via the TLR4 signaling pathway, which subsequently activated MAPK/ERK-mediated FOXO3 phosphorylation and following degradation. While the gingipains, a class of proteases produced by Pg, could effectively hydrolyze FOXO3 in the cytoplasm of macrophages. Both of them decreased the nuclear level of FOXO3, followed by the release of histone deacetylase 2 (HDAC2) from the macrophage scavenger receptor 1 (Msr1) promoter, thus promoting Msr1 transcription. This enhanced MSR1-mediated lipid uptake further amplified oxidative stress-induced necroptosis in lipid-laden macrophages. In summary, Pg exacerbates macrophage oxidative stress-dependent necroptosis, thus enlarges the atherosclerotic plaque necrotic core and ultimately promotes plaque destabilization.
Collapse
Affiliation(s)
- Xiaofei Huang
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Mengru Xie
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Yixuan Wang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Xiaofeng Lu
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Feng Mei
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Kaiwen Zhang
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Xinlong Yang
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guangjin Chen
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Ying Yin
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Guangxia Feng
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Wencheng Song
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China.
| | - Xuliang Deng
- Department of Geriatric Dentistry, NMPA Key Laboratory for Dental Materials, National Engineering Laboratory for Digital and Material, Technology of Stomatology, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, China.
| | - Songling Wang
- Laboratory of Homeostatic Medicine, School of Medicine, Southern University of Science and Technology, Shenzhen, China.
- Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China.
| | - Lili Chen
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China.
| |
Collapse
|
2
|
Hu XL, Li H, Zhang GD, Lin C, Huang P, Chen XF, Wan F, Dou CW, Ju HT. Synergistic effects and mechanism of recombinant viral vector-mediated STAT1 overexpression and STAT3 silencing on glioma U251 apoptosis. Mol Biol Rep 2025; 52:482. [PMID: 40402343 PMCID: PMC12098210 DOI: 10.1007/s11033-025-10585-1] [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: 10/16/2024] [Accepted: 05/08/2025] [Indexed: 05/23/2025]
Abstract
BACKGROUND In the present study, the synergistic effects and mechanism of recombinant viral vector-mediated co-expression plasmids stat1 and stat3-siRNA on glioma were investigated in vivo and in vitro. METHODS Co-expression plasmids for stat1/stat3-siRNA were constructed and packaged into lentivirus and adenovirus for cell and animal experiments. Real-time PCR and Western blot analyses were used to detect the expression of STAT1 and STAT3 at gene and protein levels in U251 cells. CCK-8, TUNEL, flow cytometry, and cell scratching assays were performed to detect the therapeutic effect of the co-expression plasmid stat1/stat3-siRNA on glioma in vitro. U251 glioma cells were injected into nude mice to observe therapeutic effect of stat1/stat3-siRNA.Transcriptome sequencing was utilized to further explore the possible mechanism. RESULTS Treatment of glioma cells and xenograft animal model with the co-expression plasmid stat1/stat3-siRNA led to a significant increase in STAT1 and a marked decrease in STAT3 expression at both mRNA and protein expression levels. Compared to the single-gene stat1 and stat3-siRNA groups, stat1/stat3-siRNA group demonstrated a more pronounced promoting apoptosis of U251, but cell viability and migration, as well as reduced tumor growth in nude mice were not significant. Transcriptome sequencing results indicated that the modulation of multiple nodes within the FOXO signaling pathway may represent the main mechanism by which co-expression of lenti-stat1/stat3-SiRNA induces U251 cell apoptosis. CONCLUSIONS The co-expression plasmid stat1/stat3-siRNA significantly induces apoptosis more effectively than individual stat1 and stat3-siRNA constructs. The potential mechanism involves the alternation of multiple nodes in the FOXO signaling pathway.
Collapse
Affiliation(s)
- Xin-Long Hu
- Department of General Surgery, Aerospace Center Hospital, Beijing, 100049, China
| | - Hong Li
- Department of Radiation Oncology, Peking University Cancer Hospital (Inner Mongolia Campus) & Afliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, 010020, Inner Mongolia Autonomous Region, China
| | - Guo-Dong Zhang
- Department of Neurosurgery, Affiliated Hospital of Chifeng University, Chifeng, 024000, Inner Mongolia Autonomous Region, China
| | - Chao Lin
- Department of General Surgery, Beijing Nuclear Industry Hospital, Beijing, 100045, China
| | - Ping Huang
- Department of Neurosurgery, Affiliated Hospital of Inner Mongolia Medical University, No.1, Tongdao North Road, Hohhot, Inner Mongolia Autonomous Region, China
| | - Xiu-Feng Chen
- Department of General Surgery, Aerospace Center Hospital, Beijing, 100049, China
| | - Fang Wan
- School of Life Sciences, Inner Mongolia Agricultural University, Hohhot, 010000, Inner Mongolia Autonomous Region, China
| | - Chang-Wu Dou
- Department of Neurosurgery, Affiliated Hospital of Inner Mongolia Medical University, No.1, Tongdao North Road, Hohhot, Inner Mongolia Autonomous Region, China.
| | - Hai-Tao Ju
- Department of Neurosurgery, Affiliated Hospital of Inner Mongolia Medical University, No.1, Tongdao North Road, Hohhot, Inner Mongolia Autonomous Region, China.
| |
Collapse
|
3
|
Guan S, Lin Q, Huang P, Lin K, Duan S. Identification of a novel FOXO3‑associated prognostic model in hepatocellular carcinoma. Oncol Lett 2025; 29:230. [PMID: 40114746 PMCID: PMC11925000 DOI: 10.3892/ol.2025.14976] [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: 07/04/2024] [Accepted: 02/14/2025] [Indexed: 03/22/2025] Open
Abstract
Although numerous molecular classifications are available to predict the prognosis of patients with hepatocellular carcinoma (HCC), they are still unsatisfactory. Forkhead box O3 (FOXO3) has been widely reported as a transcription factor involved in human cancers, but its role in HCC remains controversial. The present study aimed to explore the role of FOXO3 in HCC, as well as to identify biomarkers and construct prognostic models based on FOXO3. FOXO3 was highly expressed in HCC and was closely associated with poor prognosis in The Cancer Genome Atlas (the training set) and International Cancer Genome Consortium (the validation set). Subsequently, a co-expression network indicated that the red modules were closely related to FOXO3. Five key FOXO3-related genes [DEAD-box helicase 55 (DDX55), RAB10, member RAS oncogene family (RAB10), RAB7A, TATA-box binding protein associated factor, RNA polymerase I subunit B (TAF1B) and TAF3] were obtained using Cox-least absolute shrinkage and selection operator analyses. The 5-gene signature successfully predicted the prognosis of patients with HCC in both the training and validation sets. Enrichment analysis suggested marked differences in AKT and cell cycle-related (E2F targets and G2/M checkpoints) pathways between HCC subgroups. Furthermore, the tumor microenvironment analysis suggested that the difference in the distribution of M2 macrophages among various subgroups may contribute to the poor prognosis using the CIBERSORTx framework. Furthermore, the mRNA and protein expressions of DDX55, RAB10, RAB7A, TAF1B and TAF3 were found to be higher in HCC tissues compared with paracancerous tissues using RT-qPCR and western blotting. Additionally, knockdown of RAB10, RAB7A and TAF3 inhibited proliferation of Huh7 cells, assessed by a Cell Counting Kit-8 assay. In conclusion, a novel FOXO3-related model was constructed and revealed that RAB10, RAB7A and TAF3 may be potential molecular targets or biomarkers for HCC.
Collapse
Affiliation(s)
- Songmei Guan
- Department of Clinical Pharmacy, Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangzhou 524003, P.R. China
| | - Qiang Lin
- Department of Hepatobiliary Surgery, Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangzhou 524003, P.R. China
| | - Peiwu Huang
- Department of Hepatobiliary Surgery, Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangzhou 524003, P.R. China
| | - Kangqiang Lin
- Department of Hepatobiliary Surgery, Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangzhou 524003, P.R. China
| | - Shigang Duan
- Department of Hepatobiliary Surgery, Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangzhou 524003, P.R. China
| |
Collapse
|
4
|
Li Z, Xing J. Role of sirtuins in cerebral ischemia-reperfusion injury: Mechanisms and therapeutic potential. Int J Biol Macromol 2025; 310:143591. [PMID: 40300682 DOI: 10.1016/j.ijbiomac.2025.143591] [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: 03/03/2025] [Revised: 04/22/2025] [Accepted: 04/26/2025] [Indexed: 05/01/2025]
Abstract
The high incidence and mortality rate of cardiac arrest (CA) establishes it as a critical clinical challenge in emergency medicine globally. Despite continuous advances in advanced life support (ALS) technology, the prognosis for patients experiencing cardiac arrest remains poor, with cerebral ischemia and reperfusion injury (CIRI) being a significant determinant of adverse neurological outcomes and increased mortality. Sirtuins (SIRTs) are a class of highly evolutionarily conserved NAD+-dependent histone deacylenzymes capable of regulating the expression of various cytoprotective genes to play a neuroprotective role in CIRI. SIRTs mainly regulate the levels of downstream proteins such as PGC 1-α, Nrf 2, NLRP 3, FoxOs, and PINK 1 to inhibit inflammatory response, attenuate oxidative stress, improve mitochondrial dysfunction, promote angiogenesis, and inhibit apoptosis while reducing CIRI. Natural active ingredients are widely used in regulating the protein level of SIRTs in the body because of their multi-components, multi-pathway, multi-target, and minimal toxic side effects. However, these naturally active ingredients still face many challenges related to drug targeting, pharmacokinetic properties, and drug delivery. The emergence and vigorous development of new drug delivery systems, such as nanoparticles, micromilk, and exosomes, provide strong support for solving the above problems. In the context of the rapid development of molecular biology technology, non-coding RNA (NcRNA), represented by miRNA and LncRNA, offers great potential for achieving gene-level precision medicine. In the context of multidisciplinary integration, combining SIRTs proteins with biotechnology, omics technologies, artificial intelligence, and material science will strongly promote the deepening of their basic research and expand their clinical application. This review describes the major signaling pathways of targeting SIRTs to mitigate CIRI, as well as the current research status of Chinese and Western medicine and medical means for the intervention level of SIRTs. Meanwhile, the challenges and possible solutions in the clinical application of targeted drugs are summarized. In the context of medical and industrial crossover, the development direction of SIRTs in the future is discussed to provide valuable reference for basic medical researchers and clinicians to improve the clinical diagnosis and treatment effects of CIRI.
Collapse
Affiliation(s)
- Zheng Li
- Department of Emergency Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, China.
| | - Jihong Xing
- Department of Emergency Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, China.
| |
Collapse
|
5
|
Zhou B, Liu J, Jin L, Huang X. Remimazolam alleviates hepatic ischemia-reperfusion injury by activating FOXO1/3 signaling : Remimazolam alleviates hepatic ischemia reperfusion injury. BMC Gastroenterol 2025; 25:283. [PMID: 40263992 PMCID: PMC12016092 DOI: 10.1186/s12876-025-03820-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/26/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Hepatic ischemia reperfusion injury (HIRI) frequently gives rise to aggravated liver damage. Currently, there exists a diverse range of anesthetic drugs that possess protective capabilities against ischemia-reperfusion injury (IRI). Nevertheless, the specific functions and underlying mechanisms of remimazolam (RMZL) in HIRI have not been fully elucidated. METHODS HIRI models of both hepatocytes and mice were successfully established. To evaluate liver function and injury, ELISA, HE and TUNEL staining were employed. The levels of oxidative stress markers and inflammatory factors were measured using commercial kits. Cell viability and apoptosis were measured by CCK-8 and flow cytometry, respectively. The abundance of genes and proteins was determined utilizing RT-qPCR and western blot. RESULTS It was observed that RMZL administration greatly alleviated liver damage and repressed oxidative stress and inflammation in HIRI mouse models. In vitro experiments demonstrated that RMZL strongly protected LO2 cells from H/R-induced cell damage, oxidative stress, and inflammatory responses. Moreover, FOXO1 and FOXO3, which function as classic cell protection and anti-oxidative stress factors, were observed to be downregulated in liver tissue from HIRI mouse models and H/R-challenged LO2 cells. Notably, this downregulation could be reversed by the administration of RMZL. Furthermore, FOXO1 or FOXO3 knockdown abolished the protective effects of RMZL, including promoted cell survival and inhibited oxidative stress and inflammation in LO2 cells upon H/R exposure. CONCLUSION These data provided robust support for the notion that RMZL attenuated oxidative stress and inflammation to alleviate HIRI through enhancing FOXO1 and FOXO3 expressions, suggesting that RMZL holds great promise as a potential candidate anesthetic for HIRI treatment.
Collapse
Affiliation(s)
- Bo Zhou
- Department of Anesthesiology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), No. 61 Jiefang West Road, Furong District, Changsha, Hunan Province, 410000, P.R. China
| | - Jian Liu
- Department of Anesthesiology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), No. 61 Jiefang West Road, Furong District, Changsha, Hunan Province, 410000, P.R. China
| | - Lu Jin
- Department of Anesthesiology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), No. 61 Jiefang West Road, Furong District, Changsha, Hunan Province, 410000, P.R. China
| | - Xiaoling Huang
- Department of Anesthesiology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), No. 61 Jiefang West Road, Furong District, Changsha, Hunan Province, 410000, P.R. China.
| |
Collapse
|
6
|
Zhang Y, Wang H, Wang Y, Ma B. FOXO1 mediates miR-99a-5p/E2F7 to restrain breast cancer cell proliferation and induce apoptosis. BMC Cancer 2025; 25:747. [PMID: 40264026 PMCID: PMC12013025 DOI: 10.1186/s12885-025-14111-1] [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: 12/20/2024] [Accepted: 04/08/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND FOXO1 is known to act as a tumor suppressor gene in breast cancer, but its exact mechanism of action remains unclear. OBJECTIVE This study aimed to clarify how FOXO1 suppresses breast cancer cell proliferation and induces apoptosis. METHODS Breast cancer cell lines were generated with stable knockdown or overexpression of FOXO1. RT-qPCR and western blot assays were conducted to confirm transfection efficiency. CCK-8 and colony formation assays were used to assess cell proliferation, while flow cytometry measured apoptosis. The cells were subcutaneously injected into nude mice, and the volume and mass of the resulting tumors were evaluated. Immunohistochemistry was used to analyze Ki-67 expression in the tumors. A TUNEL assay examined apoptosis in the tumor cells. We performed bioinformatic analysis to identify FOXO1-targeted miRNAs and their downstream target mRNAs. RESULTS Overexpression of FOXO1 inhibited breast cancer cell proliferation and promoted apoptosis. In contrast, knockdown of FOXO1 enhanced cell proliferation and reduced apoptosis. Among the downstream miRNAs we identified, miR-99a-5p was found to be downregulated in breast cancer tissue. FOXO1 binds to the miR-99a promoter, facilitating its transcription. Inhibition of miR-99a-5p partially reversed the effects of FOXO1 overexpression on cell proliferation and apoptosis. E2F7, a target mRNA of miR-99a-5p, showed a negative correlation with FOXO1 expression in breast cancer mRNAs we screened. Silencing E2F7 partially mitigated the inhibitory effects of miR-99a-5p on proliferation and apoptosis in FOXO1-overexpressing cells. E2F7 binds to the FOXO1 promoter, thus suppressing its transcription and reducing its expression. CONCLUSION FOXO1 suppresses breast cancer cell proliferation and promotes apoptosis by enhancing the transcription and expression of miR-99a-5p, while inhibiting its target gene E2F7. E2F7, in turn, represses the transcription of FOXO1, lowering its expression.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Thyroid and Breast Surgery, the People's Hospital of Liaoning Province, Shenyang, Liaoning, 110016, China
| | - Haili Wang
- Department of Nursing, the People's Hospital of Liaoning Province, No.33, Wenyi Road, Shenhe District, Shenyang, Liaoning, 110016, China
| | - Yiliang Wang
- Department of Anesthesiology, the First Hospital of China Medical University, No.155, Nanjing North Street, Heping District, Shenyang, Liaoning, 110001, China.
| | - Bo Ma
- Department of Nursing, the People's Hospital of Liaoning Province, No.33, Wenyi Road, Shenhe District, Shenyang, Liaoning, 110016, China.
| |
Collapse
|
7
|
Zheng Q, Lin R, Li Z, Zheng Q, Xu W. Taurine is a potential therapy for rheumatoid arthritis via targeting FOXO3 through cellular senescence and autophagy. PLoS One 2025; 20:e0318311. [PMID: 40238799 PMCID: PMC12002484 DOI: 10.1371/journal.pone.0318311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 01/13/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease closely related to aging with unclear pathogenic mechanisms. This study aims to identify the biomarkers in RA, aging and autophagy using bioinformatics and machine learning and explore the binding stability of taurine to target utilizing computer-aided drug design (CADD). METHODS We identified differentially expressed genes (DEGs) for RA, then crossed with gene libraries for aging and autophagy to identify common genes (Co-genes). We performed Gene Ontology (GO), Kyoto Encyclopedia of the Genome (KEGG), and ClueGO analysis for Co-genes. The Co-genes were subjected to support vector machine-recursive feature elimination (SVM-RFE), Degree, and Betweenness algorithms to get hub genes, then verified by an artificial neural network (ANN). After continuing to perform least absolute shrinkage and selection operator (LASSO) and weighted gene co-expression network analysis (WGCNA) on Co-genes, the results were crossed with hub genes to obtain genes, which were imported into various validation sets for receiver operating characteristics (ROC) to identify key genes. We analyzed the microRNA/TF network, enriched pathways, and immune cell infiltration for key genes. The binding stability of taurine with the target protein was verified by CADD. Finally, we used Western blot for in vitro experimental verification. RESULTS We obtained 74 Co-genes enriched in RA, cellular senescence, and regulation of programmed cell death. The model prediction of hub genes works well in ANN. The key genes (MMP9, CXCL10, IL15, FOXO3) were tested in ROC with excellent efficacy. In RA, FOXO3 expression was down-regulated while MMP9, CXCL10, and IL15 expression were upregulated, and FOXO3 was negatively correlated with MMP9, CXCL10, and IL15. Two miRNAs (hsa-mir-21-5p, hsa-mir-129-2-3p) and four TFs (CTCF, KLF, FOXC1, TP53) were associated with key genes. The immune cells positively correlated with MMP9, CXCL10, and IL15 expression and negatively correlated with FOXO3 expression were Plasma cells, CD8 T cells, memory-activated CD4 T cells, and follicular helper T cells, aggregating in RA. The binding stability of taurine with FOXO3 was verified by molecular docking and molecular dynamics simulation. In vitro experiments have indicated that taurine can upregulate the expression of FOXO3 and treat RA through the FOXO3-Parkin signaling pathway. CONCLUSIONS MMP9, CXCL10, IL15, and FOXO3 are biomarkers of RA, cellular senescence, and autophagy. Taurine might be a promising drug against RA via targeting cellular senescence and autophagy through FOXO3.
Collapse
Affiliation(s)
- Qingcong Zheng
- Department of Spinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Rongjie Lin
- Department of Orthopedic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Zhechen Li
- Department of Spinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Qingzhu Zheng
- Department of Laboratory Medicine, Fujian Medical University Union Hospital, Fuzhou, China
| | - Weihong Xu
- Department of Spinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
8
|
Cao Y, He Y, Liao L, Xu L. Circular RNAs perspective: exploring the direction of immunotherapy for colorectal cancer. Front Oncol 2025; 15:1554179. [PMID: 40291917 PMCID: PMC12021614 DOI: 10.3389/fonc.2025.1554179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Circular RNAs (circRNAs) are multifaceted molecules that play a pivotal role in regulating gene expression at both transcriptional and post-transcriptional levels. Their expression is highly tissue-specific and developmentally regulated, making them critical players in various physiological processes and diseases, particularly cancer. In colorectal cancer, circRNAs exhibit significantly dysregulated expression patterns and profoundly influence disease progression through diverse molecular mechanisms. Unraveling the complex roles of circRNAs in modulating colorectal cancer immunotherapy outcomes highlights their potential as both promising biomarkers and therapeutic targets. Moving forward, advancements in circRNA-based therapeutic strategies and delivery systems are poised to transform precision medicine, enabling early colorectal cancer diagnosis and improving patient prognosis.
Collapse
Affiliation(s)
- Yanlin Cao
- Department of Pathology, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, Hunan, China
- The First Clinical Medical College of Guangdong Medical University, Zhanjiang, China
| | - Yuxing He
- Department of Medical Laboratory Medicine, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, Hunan, China
| | - Lingshan Liao
- Department of Pathology, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, Hunan, China
| | - Lixin Xu
- Neurosurgery Department, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, Hunan, China
| |
Collapse
|
9
|
Cui J, Li H, Zhang T, Lin F, Chen M, Zhang G, Feng Z. Research progress on the mechanism of curcumin anti-oxidative stress based on signaling pathway. Front Pharmacol 2025; 16:1548073. [PMID: 40260389 PMCID: PMC12009910 DOI: 10.3389/fphar.2025.1548073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 03/24/2025] [Indexed: 04/23/2025] Open
Abstract
Oxidative stress refers to an imbalance between oxidative capacity and antioxidant capacity, leading to oxidative damage to proteins, lipids, and DNA, which can result in cell senescence or death. It is closely associated with the occurrence and development of various diseases, including cardiovascular diseases, nephropathy, malignant tumors, neurodegenerative diseases, hypertension, diabetes, and inflammatory diseases. Curcumin is a natural polyphenol compound of β-diketone, which has a wide range of pharmacological activities such as anti-inflammatory, antibacterial, anti-oxidative stress, anti-tumor, anti-fibrosis, and hypolipidemic, demonstrating broad research and development value. It has a wide range of biological targets and can bind to various endogenous biomolecules. Additionally, it maintains the redox balance primarily by scavenging ROS, enhancing the activity of antioxidant enzymes, inhibiting lipid peroxidation, and chelating metal ions. This paper systematically describes the antioxidative stress mechanisms of curcumin from the perspective of signaling pathways, focusing on the Keap1-Nrf2/ARE, NF-κB, NOX, MAPK and other pathways. The study also discusses potential pathway targets and the complex crosstalk among these pathways, aiming to provide insights for further research on curcumin's antioxidant mechanisms and its clinical applications.
Collapse
Affiliation(s)
- Jie Cui
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Haonan Li
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Tianyi Zhang
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Fengli Lin
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Meiyun Chen
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Guimin Zhang
- Lunan Pharmaceutical Group Co., Ltd., Linyi, China
| | - Zhong Feng
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
- Lunan Pharmaceutical Group Co., Ltd., Linyi, China
| |
Collapse
|
10
|
Ma X, Lin Y, Zhang L, Huang Z, Zhang Y, Fu X, Li P. The dual missions of FoxO3a in inflammatory diseases: Regulation of antioxidant enzymes and involvement in programmed cell death. Int Immunopharmacol 2025; 151:114369. [PMID: 40031428 DOI: 10.1016/j.intimp.2025.114369] [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: 01/20/2025] [Revised: 02/22/2025] [Accepted: 02/23/2025] [Indexed: 03/05/2025]
Abstract
The transcription factor FoxO3a plays a crucial role in the process of cells adapting to various stress conditions. Multiple post - translational modifications and epigenetic mechanisms work together to precisely regulate the activity of FoxO3a, influencing its subcellular localization, stability, interactions with other proteins, DNA - binding affinity, and transcriptional regulatory capacity. Under different chemical signal stimuli and subcellular environments, the activation of FoxO3a triggered by oxidative stress can initiate diverse transcriptional programs, which are essential for the body to resist oxidative damage. In the development and progression of inflammatory diseases, FoxO3a exerts an important function by regulating the expression levels of antioxidant enzymes and participating in key physiological processes such as programmed cell death. This article comprehensively reviews the structural characteristics, mechanism of action of FoxO3a, as well as its functions in regulating antioxidant enzymes and programmed cell death. The aim is to deeply explore the potential of FoxO3a as a potential therapeutic target for preventing and treating damages such as inflammatory diseases caused by cellular stress.
Collapse
Affiliation(s)
- Xiangli Ma
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Yujie Lin
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Ling Zhang
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Zhenzhen Huang
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Yurong Zhang
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Xu Fu
- Key Laboratory of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Peiwu Li
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China.
| |
Collapse
|
11
|
Feng S, Xu G, Ding Q, Shi Y. Fritillaria thunbergii Miq. Extract ameliorated experimental pulmonary fibrosis partly through the PI3K/AKT/FOXO signalling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 343:119445. [PMID: 39938765 DOI: 10.1016/j.jep.2025.119445] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/14/2025] [Accepted: 02/03/2025] [Indexed: 02/14/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Pulmonary fibrosis is an irreversible lung disease with a high mortality rate. Zhebeimu (ZBM, Fritillaria thunbergii Miq.) is a Chinese medicine commonly used for the treatment of pulmonary fibrosis in China. AIM OF THE STUDY In this study, the protective effect and mechanism of ZBM extract in the treatment of pulmonary fibrosis were investigated in vivo and in vitro. MATERIALS AND METHODS The protective effect of ZBM extract was assessed using an in vivo model of bleomycin (BLM) tracheal drip and transforming growth factor-β(TGF-β1)-induced fibroblasts to simulate pulmonary fibrosis, and lung function, lung histopathological status and hydroxyproline were tested. Relevant pathways were detected using protein blotting, immunofluorescence and immunohistochemistry. RESULTS ZBM extract effectively improved lung function, inflammatory changes and fibrotic deposition in the lungs, and reduced the expression of fibroblast markers in mice. In addition, ZBM extract significantly inhibited TGF-β1-induced hyperphosphorylation of FOXO3, and simultaneously improved the low expression level of FOXO3 prototype protein and significantly reduced the phosphorylation level of PI3K-p85 and AKT1, suggesting that ZBM extract improves lung fibrosis by inhibiting the over-activation of PI3K/AKT/FOXO signalling pathway. CONCLUSION The PI3K/AKT/FOXO signalling pathway is critical for ZBM extract to improve pulmonary fibrosis.
Collapse
Affiliation(s)
- Siwen Feng
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, 518172, China.
| | - Gonghao Xu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Qi Ding
- Shenzhen Research Institute, Beijing University of Chinese Medicine, Shenzhen, 518118, China.
| | - Yuanyuan Shi
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, 518172, China.
| |
Collapse
|
12
|
Lee YJ, Kim MN, Kim EG, Park CH, Cho JY, Ko BC, Kim MJ, Kim YH, Lee SM, Kim KW, Song TW, Sohn MH. SIRT2 Regulates Apoptosis Signaling in Hyperoxic Acute Lung Injury. Lung 2025; 203:41. [PMID: 40067456 DOI: 10.1007/s00408-025-00794-7] [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: 10/16/2024] [Accepted: 02/24/2025] [Indexed: 05/13/2025]
Abstract
PURPOSE Oxygen therapy is helpful for patients with breathing difficulties; however, sustained supplementation with high-concentration oxygen can cause hyperoxic acute lung injury. Sirtuin 2 (SIRT2), a nicotinamide adenine dinucleotide (NAD+)-dependent deacetylase, has been shown to be involved in pulmonary fibrosis, apoptosis, and inflammation. Here, we elucidated the role of SIRT2 in hyperoxic acute lung injury. METHODS Wild-type (WT) mice and SIRT2-deficient (SIRT2-/-) mice were exposed to room air or hyperoxia for 72 h. Thereafter, changes in hyperoxia-induced responses were evaluated in WT and SIRT2-/- mice. RESULTS SIRT2 expression was elevated in WT mice after hyperoxic exposure. We also observed that the levels of SIRT2 were higher in tracheal aspirates from newborns with bronchopulmonary dysplasia (BPD) than in those without BPD. Hyperoxia-induced inflammation and apoptosis were more considerably attenuated in SIRT2-/- mice than in WT mice. We also observed an interaction between SIRT2 and forkhead box O3 (FOXO3), and that SIRT2 deficiency was associated with altered acetylation levels of FOXO3 and changes in the expression of its downstream targets. Further investigation of the therapeutic effect of SIRT2 showed that hyperoxic acute lung injury was alleviated when AGK2, a SIRT2 inhibitor, was administered. CONCLUSION Taken together, SIRT2 plays a critical role in the pathogenesis of hyperoxic acute lung injury by regulating apoptotic signaling. These findings indicated that SIRT2 is potentially a novel therapeutic strategy for hyperoxic acute lung injury.
Collapse
Affiliation(s)
- Yu Jin Lee
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Institute for Immunology and Immunological Diseases, Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, 50-1, Yonsei-Ro Seodaemun-Gu, Seoul, South Korea
| | - Mi Na Kim
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Institute for Immunology and Immunological Diseases, Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, 50-1, Yonsei-Ro Seodaemun-Gu, Seoul, South Korea
| | - Eun Gyul Kim
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Institute for Immunology and Immunological Diseases, Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, 50-1, Yonsei-Ro Seodaemun-Gu, Seoul, South Korea
| | - Chang Hyun Park
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Institute for Immunology and Immunological Diseases, Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, 50-1, Yonsei-Ro Seodaemun-Gu, Seoul, South Korea
| | - Joo Yeon Cho
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Institute for Immunology and Immunological Diseases, Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, 50-1, Yonsei-Ro Seodaemun-Gu, Seoul, South Korea
| | - Byung Chan Ko
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Institute for Immunology and Immunological Diseases, Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, 50-1, Yonsei-Ro Seodaemun-Gu, Seoul, South Korea
| | - Min Jung Kim
- Department of Pediatrics, Yongin Severance Hospital, Yonsei University College of Medicine, 363 Dongbaekjukjeon-Daero Giheung-Gu, Yongin, South Korea
| | - Yoon Hee Kim
- Department of Pediatrics, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Soon Min Lee
- Department of Pediatrics, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Kyung Won Kim
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Institute for Immunology and Immunological Diseases, Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, 50-1, Yonsei-Ro Seodaemun-Gu, Seoul, South Korea
| | - Tae Won Song
- Department of Pediatrics, Ilsan Paik Hospital, Inje University College of Medicine, 170 Juhwa-Ro, Ilsanseo-Gu, Goyang, 10380, South Korea
| | - Myung Hyun Sohn
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Institute for Immunology and Immunological Diseases, Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, 50-1, Yonsei-Ro Seodaemun-Gu, Seoul, South Korea.
| |
Collapse
|
13
|
Landim-Vieira M, Nieto Morales PF, ElSafty S, Kahmini AR, Ranek MJ, Solís C. The role of mechanosignaling in the control of myocardial mass. Am J Physiol Heart Circ Physiol 2025; 328:H622-H638. [PMID: 39739566 DOI: 10.1152/ajpheart.00277.2024] [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: 04/30/2024] [Revised: 12/03/2024] [Accepted: 12/17/2024] [Indexed: 01/02/2025]
Abstract
Regulation of myocardial mass is key for maintaining cardiovascular health. This review highlights the complex and regulatory relationship between mechanosignaling and myocardial mass, influenced by many internal and external factors including hemodynamic and microgravity, respectively. The heart is a dynamic organ constantly adapting to changes in workload (preload and afterload) and mechanical stress exerted on the myocardium, influencing both physiological adaptations and pathological remodeling. Mechanosignaling pathways, such as the mitogen-activated protein kinases (MAPKs) and the phosphoinositide 3-kinases and serine/threonine kinase (PI3K/Akt) pathways, mediate downstream effects on gene expression and play key roles in transducing mechanical cues into biochemical signals, thereby modulating cellular processes, including control of myocardial mass. Dysregulation of these processes can lead to pathological cardiac remodeling, such as hypertrophic cardiomyopathy. Furthermore, recent studies have highlighted the importance of protein quality control mechanisms, such as the ubiquitin-proteasome system, in settings of extreme physiological conditions that alter the heart workload such as pregnancy and microgravity. Overall, this review provides a thorough insight into how mechanical signals are converted into chemical signals to regulate myocardial mass in both healthy and diseased conditions.
Collapse
Affiliation(s)
- Maicon Landim-Vieira
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, Florida, United States
| | - Paula F Nieto Morales
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, Florida, United States
| | - Summer ElSafty
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, Florida, United States
| | - Aida Rahimi Kahmini
- Department of Health, Nutrition, and Food Science, Florida State University, Tallahassee, Florida, United States
| | - Mark J Ranek
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, Maryland, United States
| | - Christopher Solís
- Department of Health, Nutrition, and Food Science, Florida State University, Tallahassee, Florida, United States
| |
Collapse
|
14
|
Watanabe N, Sanada E, Okano A, Nogawa T, Lai NS, Mazaki Y, Muroi M, Yashiroda Y, Yoshida M, Osada H. Violaceoid F induces nuclear translocation of FOXO3a by inhibiting CRM1 via a novel mechanism and suppresses HeLa cell growth. FEBS Lett 2025; 599:755-765. [PMID: 39727141 DOI: 10.1002/1873-3468.15085] [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: 08/13/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/28/2024]
Abstract
FOXO3a is a transcription factor involved in cell growth inhibition and apoptosis. FOXO3a is localized in the cytoplasm in cancer cells, and its nuclear translocation by small molecules is expected to prevent cancer cell growth. In this study, we screened a fungal broth library in HeLa cells using fluorescently labeled FOXO3a and an AI-based imaging system. We identified violaceoid F, which translocates FOXO3a into the nucleus by inhibiting CRM1, which is responsible for nuclear protein export. Violaceoid F was observed to target the reactive cysteine of CRM1 through its α, β-epoxyketone. However, because violaceoid F did not inhibit Crm1 in fission yeast cells, it seems to target cysteine residue(s) other than Cys528 of human CRM1 which are not targeted by other known CRM1 inhibitors, indicating that violaceoid F inhibits CRM1 via a novel mechanism.
Collapse
Grants
- 21K19418 Ministry of Education, Culture, Sports, Science and Technology
- 23H04880 Ministry of Education, Culture, Sports, Science and Technology
- 23H04882 Ministry of Education, Culture, Sports, Science and Technology
- 23H04885 Ministry of Education, Culture, Sports, Science and Technology
- 23H05473 Ministry of Education, Culture, Sports, Science and Technology
- 24K08739 Ministry of Education, Culture, Sports, Science and Technology
Collapse
Affiliation(s)
- Nobumoto Watanabe
- Chemical Resource Development Research Unit, RIKEN Center for Sustainable Resource Science, Wako, Japan
- USM-RIKEN International Centre for Advanced Science (URICAS), Universiti Sains Malaysia, Penang, Malaysia
| | - Emiko Sanada
- Chemical Resource Development Research Unit, RIKEN Center for Sustainable Resource Science, Wako, Japan
| | - Akiko Okano
- Chemical Resource Development Research Unit, RIKEN Center for Sustainable Resource Science, Wako, Japan
| | - Toshihiko Nogawa
- Molecular Structure Characterization Unit, RIKEN Center for Sustainable Resource Science, Wako, Japan
| | - Ngit Shin Lai
- USM-RIKEN International Centre for Advanced Science (URICAS), Universiti Sains Malaysia, Penang, Malaysia
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia (USM), Penang, Malaysia
| | - Yui Mazaki
- Molecular Ligand Target Research Team, RIKEN Center for Sustainable Resource Science, Wako, Japan
| | - Makoto Muroi
- Chemical Resource Development Research Unit, RIKEN Center for Sustainable Resource Science, Wako, Japan
- Biomolecular Characterization Unit, RIKEN Center for Sustainable Resource Science, Wako, Japan
| | - Yoko Yashiroda
- Molecular Ligand Target Research Team, RIKEN Center for Sustainable Resource Science, Wako, Japan
| | - Minoru Yoshida
- Chemical Genomics Research Group, RIKEN Center for Sustainable Resource Science, Wako, Japan
- Office of University Professors, The University of Tokyo, Bunkyo, Japan
- Collaborative Research Institute for Innovative Microbiology (CRIIM), The University of Tokyo, Bunkyo, Japan
| | - Hiroyuki Osada
- Chemical Resource Development Research Unit, RIKEN Center for Sustainable Resource Science, Wako, Japan
- USM-RIKEN International Centre for Advanced Science (URICAS), Universiti Sains Malaysia, Penang, Malaysia
- Institute of Microbial Chemistry (BIKAKEN), Shinagawa, Japan
| |
Collapse
|
15
|
Wu S, Weng J, Pan Y, Wen Z, Zeng J, Lou Y, Tong S, Liao P, Li N, Yu Z, Xia J. Disulfiram/Cu targeting FOXO6 modulates sensitivity of hepatocellular carcinoma to lenvatinib via disrupt choline metabolic. Cell Signal 2025; 127:111563. [PMID: 39694126 DOI: 10.1016/j.cellsig.2024.111563] [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: 09/18/2024] [Revised: 11/22/2024] [Accepted: 12/09/2024] [Indexed: 12/20/2024]
Abstract
Disulfiram/Cu(DSF/Cu) has a known pharmacokinetic and safety profile, exerting a strong antitumor effect. Oral tyrosine kinase inhibitors including lenvatinib are approved as first-line therapy for treating advanced unresectable hepatocellular carcinoma (HCC). These patients still have limited survival due to drug resistance. Disulfiram/Cu and lenvatinib are the promising antitumor treatments. In this study, we studied whether Disulfiram/Cu increased lenvatinib sensitivity in HCC cells. Moreover, the potential drug targets of Disulfiram/Cu and associated mechanisms were explored. We mainly investigated Autophagic flux was determined via immunofluorescence analysis and confocal microscopy. p-PI3K, p-AKT, p62, LC3B, FOXO6, and CHKA proteins associated with autophagy were detected by immunoblotting. In addition, antitumour activity of Disulfiram/Cu in combination with lenvatinib was examined in vivo through construction of the nude mouse transplant tumor model. Furthermore, our results show disulfiram/Cu combined with lenvatinib exerted the synergistic impact on treating HCC in vitro. Mechanistically, transcriptome combined with metabolome reveals Disulfiram/Cu targeting FOXO6 induction of autophagy mediated inhibits cell growth in hepatocellular carcinoma by downregulating CHKα for inhibiting AKT pathway activation while blocking choline metabolic reprogramming in HCC. These effects mostly explain the tumor-promoting effect of FOXO6 on HCC. In general, the results illustrate the mechanistic associations between metabolites and tumor cell malignant phenotype, contributing to developing new anti-HCC pharmacological treatments by Inhibiting FOXO6 for disrupting choline metabolic pathway.
Collapse
Affiliation(s)
- Shiyi Wu
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Jialu Weng
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Yating Pan
- Department of Respiratory Medicine, Yongkang First People's Hospital, Yongkang 321300, China
| | - Zhikai Wen
- Department of Liver and Gall Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Jing Zeng
- Department of Otorhinolaryngology, Hanshou County Hospital of Traditional Chinese Medicine, Changdei 415900, China
| | - Yunwei Lou
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Songjian Tong
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Pan Liao
- The School of Medicine, Nankai University 94 Weijin Road, Tianjin 300071, China
| | - Na Li
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
| | - Zhijie Yu
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China.
| | - Jinglin Xia
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China; Liver Cancer Institute, Zhongshan Hospital of Fudan University, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China.
| |
Collapse
|
16
|
Luperchio AM, Salamango DJ. Defining the Protein Phosphatase 2A (PP2A) Subcomplexes That Regulate FoxO Transcription Factor Localization. Cells 2025; 14:342. [PMID: 40072071 PMCID: PMC11899004 DOI: 10.3390/cells14050342] [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: 01/30/2025] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/15/2025] Open
Abstract
The family of forkhead box O (FoxO) transcription factors regulate cellular processes involved in glucose metabolism, stress resistance, DNA damage repair, and tumor suppression. FoxO transactivation activity is tightly regulated by a complex network of signaling pathways and post-translational modifications. While it has been well established that phosphorylation promotes FoxO cytoplasmic retention and inactivation, the mechanism underlying dephosphorylation and nuclear translocation is less clear. Here, we investigate the role of protein phosphatase 2A (PP2A) in regulating this process. We demonstrate that PP2A and AMP-activated protein kinase (AMPK) combine to regulate nuclear translocation of multiple FoxO family members following inhibition of metabolic signaling or induction of oxidative stress. Moreover, chemical inhibitor studies indicate that nuclear accumulation of FoxO proteins occurs through inhibition of nuclear export as opposed to promoting nuclear import as previously speculated. Functional, genetic, and biochemical studies combine to identify the PP2A complexes that regulate FoxO nuclear translocation, and the binding motif required. Mutating the FoxO-PP2A interface to enhance or diminish PP2A binding alters nuclear translocation kinetics accordingly. Together, these studies shed light on the molecular mechanisms regulating FoxO nuclear translocation and provide insights into how FoxO regulation is integrated with metabolic and stress-related stimuli.
Collapse
Affiliation(s)
| | - Daniel J. Salamango
- Department of Microbiology, Immunology, and Molecular Genetics, UT Health Science Center, San Antonio, TX 78229, USA;
| |
Collapse
|
17
|
Zhang R, Kou N, Liu F, Tong H, Li S, Ren L. The Sirt1/FOXO signal pathway involves in regulating osteomyelitis progression via modulating mitochondrial dysfunctions and osteogenic differentiation. J Mol Histol 2025; 56:87. [PMID: 39939446 DOI: 10.1007/s10735-025-10370-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 02/05/2025] [Indexed: 02/14/2025]
Abstract
The Sirtuin-1 (Sirt1) gene has been reported to be closely associated with the progression of multiple diseases, but its role in regulating osteomyelitis (OM) pathogenesis has not been explored. The murine long bone-derived osteocyte-like MLO-Y4 cells and osteoblast-like MC3T3-E1 cells were exposed to Staphylococcal protein A (SpA) treatment to establish the in vitro OM models. The expression levels of Osteoblast-specific genes (OCN, OPN and RUNX2), osteoclastic genes (CTSK, MMP9 and ACP5) and the FOXO pathway-related proteins (FOXO1, p-FOXO1, FOXO3 and p-FOXO3) were detected by performing Real-Time qPCR and Western Blot analysis. Osteoblastic differentiation of the cells were evaluated by using the alizarin red S staining assay and TRAP staining assay, and membrane potential and superoxide production were measured to evaluate the mitochondrial functions of the cells. SpA treatment significantly suppressed osteogenic differentiation and induced mitochondrial dysfunction in MLO-Y4 and MC3T3-E1 cells, and promoting osteoclastogenesis in RAW264.7 cells, suggesting that the in vitro OM models were successfully established. Of note, SpA decreased the expression levels of Sirt1 in the OM cells, and SpA-induced detrimental effects on the OM cells were all reversed by overexpressing Sirt1. Mechanistically, Sirt1-overexpression increased the levels of phosphorylated FOXO-related proteins (p-FOXO1 and p-FOXO3) to activate the FOXO signal pathway and ameliorated OM progression in SpA-treated cells. Collectively, it was revealed in the present study that overexpression of Sirt1 activated the FOXO signal pathway to ameliorate SpA-induced detrimental effects in the OM cells, and Sirt1 could be potentially used as therapeutic agent for OM in clinic.
Collapse
Affiliation(s)
- Runyao Zhang
- Department of Orthopedics, Guiqian International Hospital, No. 1 Dongfeng Avenue, Wudang District, Guiyang City, Guizhou Province, People's Republic of China
| | - Nannan Kou
- Department of Traumatology, The Second Affiliated Hospital of Kunming Medical University, No. 374, Dianmian Avenue. Wuhua District, Kunming City, Yunnan Province, People's Republic of China
| | - Feifei Liu
- Department of Spine Surgery, The First Affiliated Hospital of Dali University, No. 32, Jiashibo Avenue, Dali, Yunnan Province, People's Republic of China
| | - Huan Tong
- Department of Spine Surgery, The First Affiliated Hospital of Dali University, No. 32, Jiashibo Avenue, Dali, Yunnan Province, People's Republic of China
| | - Shaobo Li
- Department of Spine Surgery, The First Affiliated Hospital of Dali University, No. 32, Jiashibo Avenue, Dali, Yunnan Province, People's Republic of China
| | - Lirong Ren
- Department of Spine Surgery, The First Affiliated Hospital of Dali University, No. 32, Jiashibo Avenue, Dali, Yunnan Province, People's Republic of China.
| |
Collapse
|
18
|
Gao T, Yan N, Pu Y, Zhang Z, Duan Z, Tang Z, Huang D, Chen Y, Yuan S, Yan X, Yuan M. Ginger leaf polyphenols mitigate β-amyloid toxicity via JNK/FOXO pathway activation in Caenorhabditis elegans. Food Funct 2025; 16:1072-1085. [PMID: 39829385 DOI: 10.1039/d4fo03238a] [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: 01/22/2025]
Abstract
β-Amyloid (Aβ) aggregation is the major pathological feature of Alzheimer's disease (AD), resulting in oxidative stress and further exacerbating Aβ aggregation. Ginger leaf polyphenols (GLP) have been found to possess antioxidant activity, evidencing their potential in addressing AD. GLP is mainly composed of 12 polyphenols, 8 organic acids, and 6 glycosides, of which polyphenols are predominantly composed of apigenin, kaempferol, and quercetin derivatives. Moreover, GLP alleviates reproductive toxicity, longevity toxicity, and neurotoxicity induced by Aβ via regulating the antioxidase system in Caenorhabditis elegans. As shown by the network pharmacology results, GLP might activate the JNK/Foxo signaling pathway to regulate the antioxidase system, which was evidenced by the up-regulation of gene expression levels of jnk-1, daf-16, sod-3, and hsp-16.2. Overall, GLP might be a potential antioxidant for combating AD.
Collapse
Affiliation(s)
- Tao Gao
- College of Life Science, Sichuan Agricultural University, Yaan 625014, China.
| | - Ningning Yan
- College of Life Science, Sichuan Agricultural University, Yaan 625014, China.
| | - Yaying Pu
- Yaan People's Hospital, Yaan, 625099, China.
| | - Zhonghao Zhang
- College of Life Science, Sichuan Agricultural University, Yaan 625014, China.
| | - Zhihao Duan
- College of Life Science, Sichuan Agricultural University, Yaan 625014, China.
| | - Zizhong Tang
- College of Life Science, Sichuan Agricultural University, Yaan 625014, China.
| | - Daojian Huang
- Dazhu County Scientific and Technical Information Institute, Dazhou, 635100, China
| | - Yanger Chen
- College of Life Science, Sichuan Agricultural University, Yaan 625014, China.
| | - Shu Yuan
- College of Resources, Sichuan Agricultural University, Chengdu, 611130, China
| | | | - Ming Yuan
- College of Life Science, Sichuan Agricultural University, Yaan 625014, China.
| |
Collapse
|
19
|
Zheng H, Li L, Wang D, Zhang S, Li W, Cheng M, Ge C, Chen J, Qiang Y, Chen F, Yu Y. FoxO is required for neoblast differentiation during planarian regeneration. Int J Biol Macromol 2025; 288:138729. [PMID: 39672403 DOI: 10.1016/j.ijbiomac.2024.138729] [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: 08/19/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 12/15/2024]
Abstract
Stem cells are of great importance in the maintenance and regeneration of tissues, with Forkhead box O (FoxO) proteins emerging as pivotal regulators of their functions. However, the precise impact of FoxO proteins on stem cell behavior within regenerative environments remains ambiguous. Planarians, renowned for their abundance of adult stem cells (neoblasts), serve as an excellent model for investigating the dynamics of stem cells during regeneration. In this study, we identified DjfoxO, a conserved foxO gene in the planarian Dugesia japonica, and demonstrated its expression in neoblasts, with elevated levels detected in the regenerative blastema during the regeneration process. Using a FoxO inhibitor (AS1842856) together with RNA interference techniques, we demonstrated that inhibition of FoxO signaling in planarians hinders the regeneration of missing tissues, including the central nervous system, eyespots, anterior intestinal branches, and pharynx. It is noteworthy that the knockdown of DjfoxO does not significantly affect the mitotic activity of neoblasts. Conversely, it impedes the production of lineage-specific progenitors, potentially via modulation of the Erk pathway. These findings elucidate the instructive function of FoxO signaling in regulating stem cell differentiation and provide valuable insights into its potential for improving stem cell-based regenerative therapies.
Collapse
Affiliation(s)
- Hanxue Zheng
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Linfeng Li
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Du Wang
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Shengchao Zhang
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Wenhui Li
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Mengdi Cheng
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Cui Ge
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Jiayi Chen
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Yanmei Qiang
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China
| | - Fulin Chen
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China; Provincial Key Laboratory of Biotechnology of Shaanxi, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, China
| | - Yuan Yu
- Laboratory of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an, China; Provincial Key Laboratory of Biotechnology of Shaanxi, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, China.
| |
Collapse
|
20
|
Zaccarelli-Magalhães J, Citadin CT, Langman J, Smith DJ, Matuguma LH, Lin HW, Udo MSB. Protein arginine methyltransferases as regulators of cellular stress. Exp Neurol 2025; 384:115060. [PMID: 39551462 PMCID: PMC11973959 DOI: 10.1016/j.expneurol.2024.115060] [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: 10/24/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/19/2024]
Abstract
Arginine modification can be a "switch" to regulate DNA transcription and a post-translational modification via methylation of a variety of cellular targets involved in signal transduction, gene transcription, DNA repair, and mRNA alterations. This consequently can turn downstream biological effectors "on" and "off". Arginine methylation is catalyzed by protein arginine methyltransferases (PRMTs 1-9) in both the nucleus and cytoplasm, and is thought to be involved in many disease processes. However, PRMTs have not been well-documented in the brain and their function as it relates to metabolism, circulation, functional learning and memory are understudied. In this review, we provide a comprehensive overview of PRMTs relevant to cellular stress, and future directions into PRMTs as therapeutic regulators in brain pathologies.
Collapse
Affiliation(s)
- Julia Zaccarelli-Magalhães
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Cristiane Teresinha Citadin
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Julia Langman
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Drew James Smith
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Luiz Henrique Matuguma
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Hung Wen Lin
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA.
| | - Mariana Sayuri Berto Udo
- Department of Neurology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA.
| |
Collapse
|
21
|
O'Mahony C, Hidalgo-Lanussa O, Barreto GE. Unveiling FOXO3's metabolic contribution to menopause and Alzheimer's disease. Exp Gerontol 2025; 200:112679. [PMID: 39778695 DOI: 10.1016/j.exger.2025.112679] [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: 07/22/2024] [Revised: 01/03/2025] [Accepted: 01/05/2025] [Indexed: 01/11/2025]
Abstract
The increasing prevalence of Alzheimer's disease (AD) calls for a comprehensive exploration of its complex etiology, with a focus on sex-specific vulnerability, particularly the heightened susceptibility observed in postmenopausal women. Neurometabolic alterations during the endocrine transition emerge as early indicators of AD pathology, including reduced glucose metabolism and increased amyloid-beta (Aβ) deposition. The fluctuating endocrine environment, marked by declining estradiol levels and reduced estrogen receptor beta (ERβ) activity, further exacerbates this process. In this context, here we explore the potential of forkhead box O3 (FOXO3) as a critical mediator linking metabolic disturbances to hormonal decline. We propose that FOXO3 plays a key role in the intersection of menopause and AD, given its dysregulation in both AD patients and postmenopausal women, modulating cellular metabolism through interactions with the AMPK/AKT/PI3K pathways. This relationship highlights the intersection between hormonal changes and increased AD susceptibility. This review aims to open a discussion on FOXO3's contribution to the metabolic dysregulation seen in menopause and its impact on the progression of AD. Understanding the functional role of FOXO3 in menopause-associated metabolic changes could lead to targeted therapeutic strategies, offering novel insights for managing for this condition.
Collapse
Affiliation(s)
- Christopher O'Mahony
- Department of Biological Sciences, University of Limerick, Limerick V94 T9PX, Ireland
| | - Oscar Hidalgo-Lanussa
- Department of Biological Sciences, University of Limerick, Limerick V94 T9PX, Ireland
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick V94 T9PX, Ireland.
| |
Collapse
|
22
|
Tong H, Zhang J, Jiang L, Qu R, Lu T, Hu J. Antiviral activity of HuaganJiedu decoction (HGJDD) against hepatitis B virus (HBV) through FOXO4/ERK/HNF4α signal pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 340:119238. [PMID: 39701219 DOI: 10.1016/j.jep.2024.119238] [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: 09/26/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 12/21/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Chronic hepatitis B virus (HBV) infection is still a widespread global health issue. HuaganJiedu Decoction (HGJDD) is a common prescription for treating HBV in China, which has the effect of enhancing antiviral efficacy and improving clinical efficacy. However, its precise mechanism of action remains unclear, warranting further investigation to elucidate its therapeutic potential and integration into standard medical practices. AIM OF THE STUDY This study aims to explore the therapeutic mechanism of HuaganJiedu Decoction (HGJDD) in HBV. MATERIALS AND METHODS We investigated the therapeutic potential of HGJDD, and LC-MS analysis characterized the chemical profile of HGJDD. In vitro, we utilized HepG2.2.15 cell line to assess cytotoxicity and treatment efficacy of HGJDD compared to Entecavir controls. In vivo, assessments included monitoring HBV-related biomarkers and viral load. Network pharmacology and RNA-seq analyses identified molecular pathways and targets influenced by HGJDD treatment. Immunofluorescence and Western blotting provided further insights into the therapeutic mechanisms underlying HGJDD for HBV. RESULTS HGJDD showed no toxicity on HepG2.2.15 cells at 10%, 20%, 40%, and 80% serum concentrations. In vitro, HGJDD reduced HBsAg, HBeAg, and HBV DNA levels by dose-dependently and time-dependently. HGJDD can decrease the levels of HBsAg, HBeAg, and HBV DNA in serum and liver levels, meanwhile the therapeutic effect of high-dose HGJDD approach to EVT's in HBV Tg mice. According to intersection of network pharmacology and transcriptome, FOXO signal pathway was highlighted as potential targets and Immunofluorescence find that FOXO4D protein expression lever was increased in three HGJDD group, especially in high-dose HGJDD group. Western blotting confirmed increased level of FOXO4, ERK, and p-ERK and decreased levels of HNF4α, which reflected that the therapeutic effect was closely to FOXO4/ERK/HNF4α signal pathway. CONCLUSIONS Traditional Chinese medicine (TCM) offers diverse herbal treatments for HBV, with HGJDD showing efficacy in reducing HBsAg, HBeAg, and HBV DNA levels at cellular and animal levels. This study identified that FOXO4/ERK/HNF4α signal pathway played an important role in HGJDD's therapeutic effects. These findings support HGJDD's potential in HBV treatment, providing a scientific basis for clinical use.
Collapse
Affiliation(s)
- Hongxuan Tong
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Jiale Zhang
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Lijie Jiang
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Rendong Qu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 10029, China
| | - Tao Lu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 10029, China
| | - Jingqing Hu
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
23
|
Min Q, Chen X, Yifei G, Baifeng S, Zichuan W, Xiaolong S, Huajiang C, Wen Y, Yang L. FOXO3a overexpression ameliorates intervertebral disc degeneration by decreasing NLRP3-mediated pyroptosis. Int Immunopharmacol 2025; 144:113596. [PMID: 39579536 DOI: 10.1016/j.intimp.2024.113596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/30/2024] [Accepted: 11/06/2024] [Indexed: 11/25/2024]
Affiliation(s)
- Qi Min
- Department of Orthopaedics, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai, China
| | - Xu Chen
- Department of Orthopaedics, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai, China
| | - Gu Yifei
- Department of Orthopaedics, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai, China
| | - Sun Baifeng
- Department of Orthopaedics, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai, China
| | - Wu Zichuan
- Department of Orthopaedics, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai, China
| | - Shen Xiaolong
- Department of Orthopaedics, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai, China
| | - Chen Huajiang
- Department of Orthopaedics, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai, China
| | - Yuan Wen
- Department of Orthopaedics, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai, China.
| | - Liu Yang
- Department of Orthopaedics, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai, China.
| |
Collapse
|
24
|
Orea-Soufi A, Dávila D, Salazar-Roa M, Lorente M, Velasco G. Phosphorylation of FOXO Proteins as a Key Mechanism to Regulate Their Activity. Methods Mol Biol 2025; 2871:11-18. [PMID: 39565574 DOI: 10.1007/978-1-0716-4217-7_2] [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: 11/21/2024]
Abstract
Phosphorylation of FOXO transcription factors is one of the key mechanisms involved in the regulation of the activity, nucleo-cytosolic shuttling, and stability of this family of proteins. Here we describe several experimental approaches allowing analysis of changes in the phosphorylation of these proteins upon exposure to different stimuli.
Collapse
Affiliation(s)
- Alba Orea-Soufi
- ABC-RI, Algarve Biomedical Center Research Institute, Algarve Biomedical Center, Faro, Portugal
- Faculty of Medicine and Biomedical Sciences, University of Algarve, Faro, Portugal
| | - David Dávila
- Department of Biochemistry and Molecular Biology, School of Biology, Complutense University, Madrid, Spain
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, Spain
| | - María Salazar-Roa
- Department of Biochemistry and Molecular Biology, School of Biology, Complutense University, Madrid, Spain
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, Spain
| | - Mar Lorente
- Department of Biochemistry and Molecular Biology, School of Biology, Complutense University, Madrid, Spain
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, Spain
| | - Guillermo Velasco
- Department of Biochemistry and Molecular Biology, School of Biology, Complutense University, Madrid, Spain.
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, Spain.
| |
Collapse
|
25
|
Amenabar C, Jimenez L, Mourato C, Mayoral-Varo V, Megías D, Ferreira BI, Link W. Multiplexed Dual-Color Fluorescence-Based Distinction Between Nuclear Trapping and Translocation of FOXO3. Methods Mol Biol 2025; 2871:163-170. [PMID: 39565587 DOI: 10.1007/978-1-0716-4217-7_15] [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: 11/21/2024]
Abstract
FOXO3 is a transcription factor that mainly exerts its functions in the cell nucleus. The amino acid sequence of FOXO3 contains a nuclear localization sequence (NLS) and a nuclear export sequence (NES) allowing for nuclear/cytoplasmic shuttling that plays an important role in regulating FOXO3 activity. Nuclear accumulation of FOXO3 proteins can be the result of translocation to the nucleus triggered by upstream regulatory input or trapping of FOXO3 within the nucleus through the inhibition of its nuclear export via the receptor CRM1. In order to distinguish these two modes of FOXO3 activation, we have generated a multiplexed assay. The development of this platform includes a reporter cell line that monitors CRM1 activity by using RFP-labeled HIV-1 Rev. protein with a strong heterologous NES. Simultaneously, the intracellular localization of FOXO3 can be monitored by a second cell line stably expressing GFP-FOXO3. Here we describe a detailed protocol on how to co-culture these reporter cell lines and use them to interrogate compound-induced FOXO3 activation in order to understand the mode of action.
Collapse
Affiliation(s)
- Carlos Amenabar
- Sols-Morreale Biomedical Research Institute (IIBM), Spanish National Research Council (CSIC), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Lucia Jimenez
- Sols-Morreale Biomedical Research Institute (IIBM), Spanish National Research Council (CSIC), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Cristiana Mourato
- ABC-RI, Algarve Biomedical Center Research Institute, Algarve Biomedical Center, Faro, Portugal
- Faculty of Medicine and Biomedical Sciences, University of Algarve, Faro, Portugal
| | - Victor Mayoral-Varo
- Sols-Morreale Biomedical Research Institute (IIBM), Spanish National Research Council (CSIC), Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Facultad HM de Ciencias de la Salud, Universidad Camilo José Cela, Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain
| | - Diego Megías
- Confocal Microscopy Unit, Biotechnology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Advanced Optical Microscopy Unit, Central Core Facilities, Instituto de Salud Carlos III, Madrid, Spain
| | - Bibiana I Ferreira
- ABC-RI, Algarve Biomedical Center Research Institute, Algarve Biomedical Center, Faro, Portugal.
- Faculty of Medicine and Biomedical Sciences, University of Algarve, Faro, Portugal.
| | - Wolfgang Link
- Department of Cancer Biology, Sols-Morreale Biomedical Research Institute (IIBM), Spanish National Research Council (CSIC), Universidad Autónoma de Madrid (UAM), Madrid, Spain.
| |
Collapse
|
26
|
Link W, Ferreira BI. FOXO Transcription Factors: A Brief Overview. Methods Mol Biol 2025; 2871:1-8. [PMID: 39565573 DOI: 10.1007/978-1-0716-4217-7_1] [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: 11/21/2024]
Abstract
Forkhead box O (FOXO) transcription factors constitute a mammalian family of proteins, comprising FOXO1, FOXO3, FOXO4, and FOXO6. Originally recognized as downstream regulators within the insulin pathway, FOXO factors exhibit the ability to bind to diverse target gene promoters, thereby governing crucial facets of cellular homeostasis. These encompass cellular energy generation, resilience against oxidative stress, and the modulation of cell viability and proliferation. The dysregulation of FOXO proteins has been established as pivotal in metabolic disorders, human longevity, and the inhibition of tumorigenesis. Notably subject to posttranslational modifications for regulation, FOXO inactivation predominantly arises from excessive activation of their upstream modifying enzymes, presenting a plethora of potential avenues for pharmaceutical reinstatement of FOXO activity.
Collapse
Affiliation(s)
- Wolfgang Link
- Department of Cancer Biology, Sols-Morreale Biomedical Research Institute (IIBM), Spanish National Research Council (CSIC), Universidad Autónoma de Madrid (UAM), Madrid, Spain.
| | - Bibiana I Ferreira
- ABC-RI, Algarve Biomedical Center Research Institute, Algarve Biomedical Center, Faro, Portugal.
- Faculty of Medicine and Biomedical Sciences, University of Algarve, Faro, Portugal.
| |
Collapse
|
27
|
Jimenez L, Domínguez L, Amenabar C, Serrão G, Link W, Ferreira BI, Santos B. Immunocytochemistry-Based Detection of FOXO Isoforms in Human Cancer and Fibroblasts. Methods Mol Biol 2025; 2871:171-176. [PMID: 39565588 DOI: 10.1007/978-1-0716-4217-7_16] [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: 11/21/2024]
Abstract
In mammals, the FOXO protein family consists of four distinct isoforms: FOXO1, FOXO3, FOXO4, and FOXO6. These isoforms are key players in a wide spectrum of physiological and pathological processes, including context-specific tumor suppression. FOXO3, in particular, has emerged as a gene associated with extraordinary human longevity. While these four FOXO isoforms share common biological functions, the mechanisms underlying their overlapping and distinct roles remain less understood. It is believed that intrinsic properties and context-dependent factors contribute to isoform-specific and nonredundant FOXO functions. One promising avenue for unraveling the commonalities and specificities of these proteins involves characterizing their expression patterns in specific cell types and their activation in response to different stimuli. To facilitate this, we have developed immunocytochemistry methods capable of detecting FOXO isoforms in a highly specific manner within various human cancer cell types and fibroblasts. Importantly, this approach enables the visualization of endogenous FOXO proteins as they translocate into the cell nucleus in response to different stimuli. In this article, we present a comprehensive guide to these procedures, offering valuable insights into the distinct roles of FOXO isoforms in cellular function.
Collapse
Affiliation(s)
- Lucia Jimenez
- Sols-Morreale Biomedical Research Institute (IIBM), Spanish National Research Council (CSIC), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Lucía Domínguez
- Sols-Morreale Biomedical Research Institute (IIBM), Spanish National Research Council (CSIC), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Carlos Amenabar
- Sols-Morreale Biomedical Research Institute (IIBM), Spanish National Research Council (CSIC), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Gisela Serrão
- ABC-RI, Algarve Biomedical Center Research Institute, Algarve Biomedical Center, Faro, Portugal
- Faculty of Medicine and Biomedical Sciences, University of Algarve, Faro, Portugal
| | - Wolfgang Link
- Department of Cancer Biology, Sols-Morreale Biomedical Research Institute (IIBM), Spanish National Research Council (CSIC), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Bibiana I Ferreira
- ABC-RI, Algarve Biomedical Center Research Institute, Algarve Biomedical Center, Faro, Portugal.
- Faculty of Medicine and Biomedical Sciences, University of Algarve, Faro, Portugal.
| | - Bruno Santos
- ABC-RI, Algarve Biomedical Center Research Institute, Algarve Biomedical Center, Faro, Portugal.
- Faculty of Medicine and Biomedical Sciences, University of Algarve, Faro, Portugal.
| |
Collapse
|
28
|
Zhu G, Li X, Gao Q, Wang Y, Li J, Huang Z, Lin Y. GYY4137 protects against type 2 diabetes mellitus-associated myocardial autophagy by suppressing FOXO1 signal pathway. Anim Cells Syst (Seoul) 2024; 29:13-23. [PMID: 39777025 PMCID: PMC11703514 DOI: 10.1080/19768354.2024.2442398] [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: 07/03/2024] [Revised: 09/19/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025] Open
Abstract
Purpose: Diabetic cardiomyopathy (DCM) is a major complication of type 2 diabetes mellitus (T2DM), but its effective prevention and treatment are still limited. We investigated the effects of GYY4137, a slow-releasing hydrogen sulfide donor, and its downstream mediator forkhead box protein O1 (FOXO1) on T2DM-associated DCM. Methods: In vivo, T2DM mice were induced by a high-fat diet coupled with streptozotocin injection. Intragastric administration of GYY4137 was also performed. In vitro, AC16 cardiomyocytes were treated with glucose and palmitate to mimic high-glucose and high-fat (HGHF) conditions, in which GYY4137 or a FOXO1 inhibitor (AS1842856) was also introduced. Bioinformatics analysis was performed using public GEO datasets. Results: GYY4137 demonstrated a protective effect against cardiac dysfunction, fibrosis, and autophagy in cardiac tissues of T2DM mice. Moreover, GYY4137 alleviated cell injury and lipid accumulation in HGHF-treated AC16 cells. In both in vivo and in vitro models, hyperactivation of autophagy was dampened by GYY4137. Bioinformatic analysis revealed the potential role of the FOXO pathway and autophagy in DCM. Further experiments showed that GYY4137 rescued diabetes-induced overexpression of FOXO1. AS1842856 displayed a notable capacity to shield cardiomyocytes against diabetes-induced injury similar to that achieved by GYY4137. Conclusion: GYY4137 protected against cardiac dysfunction and fibrosis in T2DM mice, and the mechanism might involve suppression of FOXO1-induced autophagy.
Collapse
Affiliation(s)
- Gaofeng Zhu
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, People’s Republic of China
| | - Xiaoyong Li
- Department of General Surgery, Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Qinyuan Gao
- Yunkang School of Medicine and Health, Nanfang College, Guangzhou, People’s Republic of China
| | - Yuanjun Wang
- Yunkang School of Medicine and Health, Nanfang College, Guangzhou, People’s Republic of China
| | - Jiajie Li
- Yunkang School of Medicine and Health, Nanfang College, Guangzhou, People’s Republic of China
| | - Zena Huang
- Yunkang School of Medicine and Health, Nanfang College, Guangzhou, People’s Republic of China
| | - Yan Lin
- Yunkang School of Medicine and Health, Nanfang College, Guangzhou, People’s Republic of China
| |
Collapse
|
29
|
Feng Y, Xin Y, Tang W, Zhang P, Jiang Y, Li H, Gong Y, Chen F, Xu Z, Liu Z, Gao L. Repeat administration of human umbilical cord mesenchymal stem cells improves left ventricular diastolic function in mice with heart failure with preserved ejection fraction. Biochem Biophys Res Commun 2024; 737:150525. [PMID: 39142139 DOI: 10.1016/j.bbrc.2024.150525] [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: 07/25/2024] [Accepted: 08/07/2024] [Indexed: 08/16/2024]
Abstract
Currently, no therapy is proven to effectively improve heart failure with preserved ejection fraction (HFpEF). Although stem cell therapy has demonstrated promising results in treating ischemic heart disease, the effectiveness of treating HFpEF with human umbilical cord mesenchymal stem cells (hucMSCs) remains unclear. To answer this question, we administered hucMSCs intravenously (i.v.), either once or repetitively, in a mouse model of HFpEF induced by a high-fat diet and NG-nitroarginine methyl ester hydrochloride. hucMSC treatment improved left ventricular diastolic dysfunction, reduced heart weight and pulmonary edema, and attenuated cardiac modeling (inflammation, interstitial fibrosis, and hypertrophy) in HFpEF mice. Repeat hucMSC administration had better outcomes than a single injection. In vitro, hucMSC culture supernatants reduced maladaptive remodeling in neonatal-rat cardiomyocytes. Ribonucleic acid sequencing and protein level analysis of left ventricle (LV) tissues suggested that hucMSCs activated the protein kinase B (Akt)/forkhead box protein O1 (FoxO1) signaling pathway to treat HFpEF. Inhibition of this pathway reversed the efficacy of hucMSC treatment. In conclusion, these findings indicated that hucMSCs could be a viable therapeutic option for HFpEF.
Collapse
Affiliation(s)
- Yunzhen Feng
- Department of Cardiovascular Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Yuanfeng Xin
- Department of Cardiovascular Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Wenjie Tang
- Research Institute of Heart Failure, Research Center for Translational Medicine & Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200123, China
| | - Pengfei Zhang
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200123, China
| | - Yun Jiang
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200123, China
| | - Hao Li
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200123, China
| | - Yanshan Gong
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200123, China
| | - Feng Chen
- Department of Gastrointestinal Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200123, China
| | - Zhifeng Xu
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Zhongmin Liu
- Department of Cardiovascular Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China; Research Institute of Heart Failure, Research Center for Translational Medicine & Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200123, China; Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200123, China.
| | - Ling Gao
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200123, China.
| |
Collapse
|
30
|
Li S, Wan J, Peng Z, Huang Q, He B. New insights of DsbA-L in the pathogenesis of metabolic diseases. Mol Cell Biochem 2024; 479:3293-3303. [PMID: 38430301 DOI: 10.1007/s11010-024-04964-8] [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: 11/29/2023] [Accepted: 02/10/2024] [Indexed: 03/03/2024]
Abstract
Metabolic diseases, such as obesity, diabetes mellitus, and non-alcoholic fatty liver disease (NAFLD), are abnormal conditions that result from disturbances of metabolism. With the improvement of living conditions, the morbidity and mortality rates of metabolic diseases are steadily rising, posing a significant threat to human health worldwide. Therefore, identifying novel effective targets for metabolic diseases is crucial. Accumulating evidence has indicated that disulfide bond A oxidoreductase-like protein (DsbA-L) delays the development of metabolic diseases. However, the underlying mechanisms of DsbA-L in metabolic diseases remain unclear. In this review, we will discuss the roles of DsbA-L in the pathogenesis of metabolic diseases, including obesity, diabetes mellitus, and NAFLD, and highlight the potential mechanisms. These findings suggest that DsbA-L might provide a novel therapeutic strategy for metabolic diseases.
Collapse
Affiliation(s)
- Siqi Li
- Department of Geriatric Respiratory and Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jinfa Wan
- Department of Emergency Medicine, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Zhenyu Peng
- Department of Emergency Medicine, Second Xiangya Hospital, Central South University, Changsha, 410011, China
- Emergency Medicine and Difficult Diseases Institute, Central South University, Changsha, 410011, China
| | - Qiong Huang
- Department of Geriatric Respiratory and Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Baimei He
- Department of Geriatric Respiratory and Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
31
|
Zhang T, Li L, Mo X, Xie S, Liu S, Zhao N, Zhang H, Chen S, Zeng X, Wang S, Deng W, Tang Q. Matairesinol blunts adverse cardiac remodeling and heart failure induced by pressure overload by regulating Prdx1 and PI3K/AKT/FOXO1 signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156054. [PMID: 39306883 DOI: 10.1016/j.phymed.2024.156054] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 08/20/2024] [Accepted: 09/13/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND Pathological cardiac remodeling is a critical process leading to heart failure, characterized primarily by inflammation and apoptosis. Matairesinol (Mat), a key chemical component of Podocarpus macrophyllus resin, exhibits a wide range of pharmacological activities, including anti-hydatid, antioxidant, antitumor, and anti-inflammatory effects. PURPOSE This study aims to investigate whether Matairesinol alleviate cardiac hypertrophy and remodeling caused by pressure overload and to elucidate its mechanism of action. METHODS An in vitro pressure loading model was established using neonatal rat cardiomyocytes treated with angiotensin Ⅱ, while an in vivo model was created using C57 mice subjected to transverse aortic constriction (TAC). To activate the PI3K/Akt/FoxO1 pathway, Ys-49 was employed. Moreover, small interfering RNA (siRNA) and short hairpin RNA (shRNA) were utilized to silence Prdx1 expression both in vitro and in vivo. Various techniques, including echocardiography, wheat germ agglutinin (WGA) staining, HE staining, PSR staining, and Masson trichrome staining, were used to assess cardiac function, cardiomyocyte cross-sectional area, and fibrosis levels in rats. Apoptosis in myocardial tissue and in vitro was detected by TUNEL assay, while reactive oxygen species (ROS) content in tissues and cells was measured using DHE staining. Furthermore, the affinity of Prdx1 with Mat and PI3K was analyzed using computer-simulated molecular docking. Western blotting and RT-PCR were utilized to evaluate Prdx1 levels and proteins related to apoptosis and oxidative stress, as well as the mRNA levels of cardiac hypertrophy and fibrosis-related indicators. RESULTS Mat significantly alleviated cardiac hypertrophy and fibrosis induced by TAC, preserved cardiac function, and markedly reduced cardiomyocyte apoptosis and oxidative damage. In vitro, mat attenuated ang Ⅱ - induced hypertrophy of nrvms and activation of neonatal rat fibroblasts. Notably, activation of the PI3K/Akt/FoxO1 pathway and downregulation of Prdx1 expression were observed in TAC mice; however, these effects were reversed by Mat treatment. Furthermore, Prdx1 knockdown activated the PI3K/Akt/FoxO1 pathway, leading to exacerbation of the disease. Molecular docking indicated that Molecular docking indicated that Mat upregulated Prdx1 expression by binding to it, thereby inhibiting the PI3K/Akt/FoxO1 pathway and protecting the heart by restoring Prdx1 expression levels. CONCLUSION Matairesinol alleviates pressure overload-induced cardiac remodeling both in vivo and in vitro by upregulating Prdx1 expression and inhibiting the PI3K/Akt/FoxO1 pathway. This study highlights the therapeutic potential of Matairesinol in the treatment of cardiac hypertrophy and remodeling, providing a promising avenue for future research and clinical application.
Collapse
Key Words
- ANP, atrial natriuretic peptide
- Abbreviations: MAT, matairesinol
- BNP, B-type natriuretic peptide
- Cardiac fibrosis
- Cardiac hypertrophy
- Cardiac remodeling
- LV, left ventricular
- LVEDd, left ventricular end-diastolic dimension
- LVEF, left ventricular ejection fraction
- Matairesinol
- NRCFS, neonatal rat cardiac fibroblasts
- PRDX 1
- PRDX1, peroxiredoxin 1
- ROS, reactive oxygen species
- Sh-RNA, short-hairpin RNA
- Si-RNA, small interfering RNA
- TAC, transverse aortic contraction
- β-MHC, Β-myosin heavy chain
Collapse
Affiliation(s)
- Tong Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Lanlan Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Xiaotong Mo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Saiyang Xie
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Shiqiang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Nan Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Heng Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Si Chen
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, PR China
| | - Xiaofeng Zeng
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, PR China
| | - Shasha Wang
- Cardiovascular Research Institute of Wuhan University, Wuhan 430060, PR China
| | - Wei Deng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Qizhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China.
| |
Collapse
|
32
|
Negrón-Piñeiro LJ, Di Gregorio A. Single-cell Transcriptomic Studies Unveil Potential Nodes of the Notochord Gene Regulatory Network. Integr Comp Biol 2024; 64:1194-1213. [PMID: 38914463 PMCID: PMC11579531 DOI: 10.1093/icb/icae084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 06/01/2024] [Accepted: 06/16/2024] [Indexed: 06/26/2024] Open
Abstract
Transcription factors (TFs) are DNA-binding proteins able to modulate the timing, location, and levels of gene expression by binding to regulatory DNA regions. Therefore, the repertoire of TFs present in the genome of a multicellular organism and the expression of variable constellations of TFs in different cellular cohorts determine the distinctive characteristics of developing tissues and organs. The information on tissue-specific assortments of TFs, their cross-regulatory interactions, and the genes/regulatory regions targeted by each TF is summarized in gene regulatory networks (GRNs), which provide genetic blueprints for the specification, development, and differentiation of multicellular structures. In this study, we review recent transcriptomic studies focused on the complement of TFs expressed in the notochord, a distinctive feature of all chordates. We analyzed notochord-specific datasets available from organisms representative of the three chordate subphyla, and highlighted lineage-specific variations in the suite of TFs expressed in their notochord. We framed the resulting findings within a provisional evolutionary scenario, which allows the formulation of hypotheses on the genetic/genomic changes that sculpted the structure and function of the notochord on an evolutionary scale.
Collapse
Affiliation(s)
- Lenny J Negrón-Piñeiro
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA
| | - Anna Di Gregorio
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA
| |
Collapse
|
33
|
Heberle BA, Fox KL, Libermann LL, Xavier SRM, Dallarosa GT, Santos RC, Fardo DW, Viola TW, Ebbert MTW. Systematic review and meta-analysis of bulk RNAseq studies in human Alzheimer's disease brain tissue. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.07.622520. [PMID: 39574617 PMCID: PMC11580990 DOI: 10.1101/2024.11.07.622520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/12/2025]
Abstract
Objective To systematically review and meta-analyze bulk RNA sequencing studies comparing Alzheimer's disease (AD) patients with controls in human brain tissue, assessing study quality and identifying key genes and pathways. Methods We searched PubMed, Web of Science, and Scopus on September 23, 2023, for studies using bulk RNAseq on primary human brain tissue from AD patients and controls. Excluded were non-primary tissue, re-analyses without new data, limited RNA types and gene panels. Quality was assessed with a 10-category tool. Meta-analysis used high-quality datasets. Results From 3,266 records, 24 studies met criteria. Meta-analysis found 571 differentially expressed genes (DEGs) in temporal lobe and 189 in frontal lobe; overlapping pathways included "Tube morphogenesis" and "Neuroactive ligand-receptor interaction." Limitations Study heterogeneity and limited data tables constrained the review. Conclusions Rigorous methods are vital in AD transcriptomic studies. Findings enhance understanding of transcriptomic changes, aiding biomarker and therapeutic development. Registration PROSPERO (CRD42023466522).
Collapse
Affiliation(s)
- Bernardo Aguzzoli Heberle
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY
| | - Kristin L Fox
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY
- Division of Laboratory Animal Resources, University of Kentucky, Lexington, KY
| | - Lucas Lobraico Libermann
- School of Medicine, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | | | - Guilherme Tarnowski Dallarosa
- School of Medicine, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Rhaná Carolina Santos
- School of Medicine, University of the Sinos Valley (UNISINOS), Porto Alegre, RS, Brazil
| | - David W Fardo
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY
- Department of Biostatistics, University of Kentucky, Lexington, Kentucky, USA
| | - Thiago Wendt Viola
- School of Medicine, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Mark T W Ebbert
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY
- Division of Biomedical Informatics, Internal Medicine, College of Medicine, University of Kentucky, Lexington, KY
| |
Collapse
|
34
|
Hu J, Lei H, Chen J, Liu L, Gui Y, Sun K, Xu D. Aerobic exercise alleviates statin-induced PCSK9 upregulation by increasing epoxyeicosatrienoic acid levels through the FoxO3a-Sirt6 axis. JOURNAL OF SPORT AND HEALTH SCIENCE 2024; 14:101007. [PMID: 39510317 PMCID: PMC11863326 DOI: 10.1016/j.jshs.2024.101007] [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: 04/21/2024] [Revised: 08/03/2024] [Accepted: 08/30/2024] [Indexed: 11/15/2024]
Abstract
BACKGROUND Statins are the cornerstone of low-density lipoprotein cholesterol (LDL-C)-lowering therapy; however, the therapeutic efficacy of statins in countering atherosclerotic cardiovascular disease (ASCVD) is compromised by the concurrent elevation of proprotein convertase subtilisin/kexin type 9 (PCSK9), a pivotal molecule that increases LDL-C levels. Aerobic exercise lowers PCSK9 levels, but the underlying mechanism remains unclear. Therefore, we investigated how aerobic exercise can ameliorate statin-induced increases in PCSK9 levels. METHODS Three-week-old male American Institute of Cancer Research (ICR) mice were fed a high-fat-cholesterol diet (HFD) for 12 weeks and then administered atorvastatin alone or atorvastatin combined with aerobic exercise (Statin + Ex). Moreover, a total of 165 participants with stable coronary heart disease (CHD) enrolled at the Inpatient and Outpatient Departments of the Second Xiangya Hospital of Central South University, China, from January 2018 to July 2020 were randomized into the Statin group (male/female = 51/33) and Statin + Ex group (male/female = 52/29). Patients in the Statin + Ex group underwent treadmill exercise of 45-60 min/day for 7 days. RESULTS Aerobic exercise effectively alleviated statin-induced PCSK9 upregulation in human patients with CHD and hypercholesterolemic ICR mice (all p < 0.05). Mechanistically, our findings revealed that aerobic exercise induced elevated epoxyeicosatrienoic acids (EETs) plasma levels while concurrently reducing the activity of soluble epoxide hydrolase (sEH) (all p < 0.05), an enzyme responsible for EETs degradation. Further, EETs significantly suppressed PCSK9 expression, subsequently reducing the LDL-C levels (all p < 0.05); this effect was mediated via the activation of the forkhead box O3a-silent mating type information regulation 2 homolog 6 (FoxO3a-Sirt6) axis, with no impact on the sterol regulatory element binding protein 2 and 3-hydroxy-3-methylglutaryl-CoA reductase (SREBP2-HMGCR) pathway. CONCLUSION Our study sheds light on the paradigm of "Exercise is Medicine", providing evidence to support the use of statins combined with exercise in reducing LDL-C levels, and unveils potential avenues for clinical applications of sEH inhibitors, presenting novel prospects for therapeutic interventions in ASCVD.
Collapse
Affiliation(s)
- Jiahui Hu
- Department of Internal Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China; National Clinical Research Center for Metabolic Diseases, Ministry of Education, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Hao Lei
- Department of Internal Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China; Department of State Drug Clinical Trial Agency, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Jingyuan Chen
- Department of Internal Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China; National Clinical Research Center for Metabolic Diseases, Ministry of Education, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Leiling Liu
- Department of Internal Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China; National Clinical Research Center for Metabolic Diseases, Ministry of Education, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Yajun Gui
- Department of Internal Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China; Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Kaijun Sun
- Department of Internal Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China; Weifang People's Hospital, The First Affiliated Hospital of Shandong Second Medical University, Weifang 261000, China
| | - Danyan Xu
- Department of Internal Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China; National Clinical Research Center for Metabolic Diseases, Ministry of Education, The Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| |
Collapse
|
35
|
Saber S, Abdelhady R, Elhemely MA, Elmorsy EA, Hamad RS, Abdel-Reheim MA, El-Kott AF, AlShehri MA, Morsy K, AlSheri AS, Youssef ME. PU-H71 (NSC 750424): a molecular masterpiece that targets HSP90 in cancer and beyond. Front Pharmacol 2024; 15:1475998. [PMID: 39564119 PMCID: PMC11573589 DOI: 10.3389/fphar.2024.1475998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 10/22/2024] [Indexed: 11/21/2024] Open
Abstract
Heat shock protein 90 (HSP90) is a pivotal molecular chaperone with multifaceted roles in cellular health and disease. Herein, we explore how HSP90 orchestrates cellular stress responses, particularly through its partnership with heat shock factor 1 (HSF-1). PU-H71, a selective inhibitor of HSP90, demonstrates significant potential in cancer therapy by targeting a wide array of oncogenic pathways. By inducing the degradation of multiple client proteins, PU-H71 disrupts critical signaling pathways such as MAPK, PI3K/Akt, JAK/STAT, EGFR, and mTOR, which are essential for cancer cell survival, proliferation, and metastasis. We examined its impact on combating triple-negative breast cancer and enhancing the effectiveness of carbon-ion beam therapy, offering new avenues for cancer treatment. Furthermore, the dual inhibition of HSP90A and HSP90B1 by PU-H71 proves highly effective in the context of myeloma, providing fresh hope for patients with this challenging malignancy. We delve into its potential to induce apoptosis in B-cell lymphomas that rely on Bcl6 for survival, highlighting its relevance in the realm of hematologic cancers. Shifting our focus to hepatocellular carcinoma, we explore innovative approaches to chemotherapy. Moreover, the current review elucidates the potential capacity of PU-H71 to suppress glial cell activation paving the way for developing novel therapeutic strategies for neuroinflammatory disorders. Additionally, the present report also suggests the promising role of PU-H71 in JAK2-dependent myeloproliferative neoplasms. Eventually, our report sheds more light on the multiple functions of HSP90 protein as well as the potential therapeutic benefit of its selective inhibitor PU-H71 in the context of an array of diseases, laying the foundations for the development of novel therapeutic approaches that could achieve better treatment outcomes.
Collapse
Affiliation(s)
- Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| | - Rasha Abdelhady
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Fayoum University, Fayoum, Egypt
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Egyptian Chinese University, Cairo, Egypt
| | - Mai A Elhemely
- School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Elsayed A Elmorsy
- Department of Pharmacology and Therapeutics, College of Medicine, Qassim University, Buraidah, Saudi Arabia
- Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Rabab S Hamad
- Biological Sciences Department, College of Science, King Faisal University, Al Ahsa, Saudi Arabia
- Central Laboratory, Theodor Bilharz Research Institute, Giza, Egypt
| | - Mustafa Ahmed Abdel-Reheim
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Shaqra, Saudi Arabia
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni Suef, Egypt
| | - Attalla F El-Kott
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia
- Department of Zoology, Faculty of Science, Damanhour University, Damanhour, Egypt
| | - Mohammed A AlShehri
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia
| | - Kareem Morsy
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia
- Department of Zoology, Faculty of Science, Cairo University, Cairo, Egypt
| | - Ali S AlSheri
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia
| | - Mahmoud E Youssef
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| |
Collapse
|
36
|
Chen L, Zhang J, Ding L, Gu T, Andoh V, Ma A, Yao C. Transcriptomics analyses combined with intestinal microorganism survey suggest Resveratrol (RSV) anti-aging and anti-oxidant effects in silkworm (Bombyx mori). COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2024; 52:101344. [PMID: 39426068 DOI: 10.1016/j.cbd.2024.101344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/11/2024] [Accepted: 10/12/2024] [Indexed: 10/21/2024]
Abstract
The growing elderly population presents a significant concern, with the prolongation of life expectancy, aging diseases are becoming increasingly common. Resveratrol (RSV) has emerged as a promising compound for disease prevention. However, the effect of RSV on lifespan extension in different organisms, particularly the model organism silkworm, remains inconsistent. We conducted aging experiments using silkworm (B. mori) and employed transcriptomics to investigate the therapeutic effects of RSV on lifespan extension and healthy lifespan in silkworms. RSV increased the survival rate by 8.57 %-12.12 % and enhanced the antioxidant capacity of silkworms. Transcriptomic analysis demonstrated that genes in signaling pathways such as AMPK and FoxO were significantly upregulated. 16SrRNA sequencing of gut contents showed an increase in beneficial bacterial strains under the action of RSV. This study aims to enhance our understanding of lifespan regulation mechanisms using the silkworm model and provide new targets for anti-aging antioxidants research to delay the onset of age-related diseases.
Collapse
Affiliation(s)
- Liang Chen
- School of Life Sciences, Jiangsu University, 212013 Zhenjiang, China.
| | - Jiaxin Zhang
- School of Life Sciences, Jiangsu University, 212013 Zhenjiang, China
| | - Lei Ding
- School of Life Sciences, Jiangsu University, 212013 Zhenjiang, China
| | - Tongyu Gu
- School of Life Sciences, Jiangsu University, 212013 Zhenjiang, China
| | - Vivian Andoh
- School of Life Sciences, Jiangsu University, 212013 Zhenjiang, China
| | - Aiqin Ma
- Qingdao Vland Animal Health Group Co., Ltd., Qingdao 266100, China
| | - Chun Yao
- Department of Stomatology, Zhenjiang First People's Hospital, Department of Stomatology, People's Hospital Affiliated to Jiangsu University, Zhenjiang 212002, China.
| |
Collapse
|
37
|
Wang Z, Luo W, Zhao C, Yu M, Li H, Zhou F, Wang D, Bai F, Chen T, Xiong Y, Wu Y. FoxO1-modulated macrophage polarization regulates osteogenesis via PPAR-γ signaling. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167333. [PMID: 38960054 DOI: 10.1016/j.bbadis.2024.167333] [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: 01/07/2024] [Revised: 06/06/2024] [Accepted: 06/27/2024] [Indexed: 07/05/2024]
Abstract
Periodontitis, a common chronic inflammatory disease, epitomizes a significant impairment in the host immune system and an imbalance of bone metabolism. Macrophage polarization, a dynamic process dictated by the microenvironment, intricately contributes to the interplay between the immune system and bone remodeling, namely the osteoimmune system. Forkhead box protein O1 (FoxO1) has been shown to play a dramatic role in mediating oxidative stress, bone mass, as well as cellular metabolism. Nevertheless, the function and underlying mechanisms of FoxO1 in regulating macrophage polarization-mediated osteogenesis in periodontitis remain to be further elucidated. Here, we found that FoxO1 expression was closely linked to periodontitis, accompanied by aggravated inflammation. Notably, FoxO1 knockdown skewed macrophage polarization from M1 to the antiinflammatory M2 phenotype under inflammatory conditions, which rescued the impaired osteogenic potential. Mechanistically, we revealed that the enhancement of the transcription of peroxisome proliferator-activated receptor (PPAR) signaling in FoxO1-knockdown macrophages. In agreement with this contention, GW9662, a specific inhibitor of PPAR-γ signaling, greatly aggravated macrophage polarization from M2 to the M1 phenotype and attenuated osteogenic potential under inflammatory conditions. Additionally, PPAR-γ signaling agonist rosiglitazone (RSG) was applied to address ligature-induced periodontitis with attenuated inflammation. Our data lend conceptual credence to the function of FoxO1 in mediating macrophage polarization-regulated osteogenesis which serves as a novel therapeutic target for periodontitis.
Collapse
Affiliation(s)
- Zhanqi Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Wenxin Luo
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Chengzhi Zhao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Muqiao Yu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Haiyun Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Feng Zhou
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Dongyang Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Fuwei Bai
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Tao Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yi Xiong
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yingying Wu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
38
|
Zang S, Wang R, Liu Y, Zhao S, Su L, Dai X, Chen H, Yin Z, Zheng L, Liu Q, Zhai Y. Insulin Signaling Pathway Mediates FoxO-Pepck Axis Regulation of Glucose Homeostasis in Drosophila suzukii. Int J Mol Sci 2024; 25:10441. [PMID: 39408770 PMCID: PMC11482478 DOI: 10.3390/ijms251910441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/19/2024] [Accepted: 09/25/2024] [Indexed: 10/19/2024] Open
Abstract
The agricultural pest Drosophila suzukii exhibits a strong preference for feeding on fresh fruits, demonstrating high adaptability to sugary environments. Meanwhile, high sugar levels stimulate insulin secretion, thereby regulating the steady state of sugar metabolism. Understanding the mechanisms related to sugar metabolism in D. suzukii is crucial due to its adaptation to these specific environmental conditions. The insulin signaling pathway is an evolutionarily conserved phosphorylation cascade with significant roles in development and metabolism. We observed that the activation of the insulin signaling pathway inhibited FoxO activity and downregulated the expression of Pepck, thereby activating glycolysis and reducing glucose levels. By contrast, inhibiting insulin signaling increased the FoxO activity and upregulated the expression of Pepck, which activated gluconeogenesis and led to increased glucose levels. Our findings demonstrated the crucial role of the insulin signaling pathway in mediating glucose metabolism through the FoxO-Pepck axis, which supports the ecological adaptation of D. suzukii to high-sugar niches, thereby providing insights into its metabolic control and suggesting potential strategies for pest management. Elucidating these molecular processes is important for understanding metabolic regulation and ecological specialization in D. suzukii.
Collapse
Affiliation(s)
- Shuting Zang
- Institute of Plant Protection, Shandong Academy of Agricultural Sciences, Jinan 250100, China
- Key Laboratory of Natural Enemies Insects, Ministry of Agriculture and Rural Affairs, Jinan 250100, China
- College of Life Sciences, Shandong Agricultural University, Tai’an 271000, China
| | - Ruijuan Wang
- Institute of Plant Protection, Shandong Academy of Agricultural Sciences, Jinan 250100, China
- Key Laboratory of Natural Enemies Insects, Ministry of Agriculture and Rural Affairs, Jinan 250100, China
| | - Yan Liu
- Institute of Plant Protection, Shandong Academy of Agricultural Sciences, Jinan 250100, China
- Key Laboratory of Natural Enemies Insects, Ministry of Agriculture and Rural Affairs, Jinan 250100, China
| | - Shan Zhao
- Institute of Plant Protection, Shandong Academy of Agricultural Sciences, Jinan 250100, China
- Key Laboratory of Natural Enemies Insects, Ministry of Agriculture and Rural Affairs, Jinan 250100, China
| | - Long Su
- Institute of Plant Protection, Shandong Academy of Agricultural Sciences, Jinan 250100, China
- Key Laboratory of Natural Enemies Insects, Ministry of Agriculture and Rural Affairs, Jinan 250100, China
| | - Xiaoyan Dai
- Institute of Plant Protection, Shandong Academy of Agricultural Sciences, Jinan 250100, China
- Key Laboratory of Natural Enemies Insects, Ministry of Agriculture and Rural Affairs, Jinan 250100, China
| | - Hao Chen
- Institute of Plant Protection, Shandong Academy of Agricultural Sciences, Jinan 250100, China
- Key Laboratory of Natural Enemies Insects, Ministry of Agriculture and Rural Affairs, Jinan 250100, China
| | - Zhenjuan Yin
- Institute of Plant Protection, Shandong Academy of Agricultural Sciences, Jinan 250100, China
- Key Laboratory of Natural Enemies Insects, Ministry of Agriculture and Rural Affairs, Jinan 250100, China
| | - Li Zheng
- Institute of Plant Protection, Shandong Academy of Agricultural Sciences, Jinan 250100, China
- Key Laboratory of Natural Enemies Insects, Ministry of Agriculture and Rural Affairs, Jinan 250100, China
| | - Qingxin Liu
- College of Life Sciences, Shandong Agricultural University, Tai’an 271000, China
| | - Yifan Zhai
- Institute of Plant Protection, Shandong Academy of Agricultural Sciences, Jinan 250100, China
- Key Laboratory of Natural Enemies Insects, Ministry of Agriculture and Rural Affairs, Jinan 250100, China
| |
Collapse
|
39
|
Wei X, Xiong X, Wang P, Zhang S, Peng D. SIRT1-mediated deacetylation of FOXO3 enhances mitophagy and drives hormone resistance in endometrial cancer. Mol Med 2024; 30:147. [PMID: 39266959 PMCID: PMC11391609 DOI: 10.1186/s10020-024-00915-7] [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: 03/28/2024] [Accepted: 08/28/2024] [Indexed: 09/14/2024] Open
Abstract
BACKGROUND The complex interplay between Sirtuin 1 (SIRT1) and FOXO3 in endometrial cancer (EC) remains understudied. This research aims to unravel the interactions of deacetylase SIRT1 and transcription factor FOXO3 in EC, focusing on their impact on mitophagy and hormone resistance. METHODS High-throughput sequencing, cell experiments, and bioinformatics tools were employed to investigate the roles and interactions of SIRT1 and FOXO3 in EC. Co-immunoprecipitation (Co-IP) assay was used to assess the interaction between SIRT1 and FOXO3 in RL95-2 cells. Functional assays were used to assess cell viability, proliferation, migration, invasion, apoptosis, and the expression of related genes and proteins. A mouse model of EC was established to evaluate tumor growth and hormone resistance under different interventions. Immunohistochemistry and TUNEL assays were used to assess protein expression and apoptosis in tumor tissues. RESULTS High-throughput transcriptome sequencing revealed a close association between SIRT1, FOXO3, and EC development. Co-IP showed a protein-protein interaction between SIRT1 and FOXO3. Overexpression of SIRT1 enhanced FOXO3 deacetylation and activity, promoting BNIP3 transcription and PINK1/Parkin-mediated mitophagy, which in turn promoted cell proliferation, migration, invasion, and inhibited apoptosis in vitro, as well as increased tumor growth and hormone resistance in vivo. These findings highlighted SIRT1 as an upstream regulator and potential therapeutic target in EC. CONCLUSION This study reveals a novel molecular mechanism underlying the functional relevance of SIRT1 in regulating mitophagy and hormone resistance through the deacetylation of FOXO3 in EC, thereby providing valuable insights for new therapeutic strategies.
Collapse
Affiliation(s)
- Xuehua Wei
- Obstetrics and Gynecology Center, Department of Gynecology, Zhujiang Hospital, Southern Medical University, No. 253, Industry Avenue, Haizhu District, Guangzhou, 510280, Guangdong, China
| | - Xiangpeng Xiong
- Department of Gynecology, Jiangxi Maternal and Child Health Hospital, Nanchang, 336000, China
| | - Pingping Wang
- Obstetrics and Gynecology Center, Department of Gynecology, Zhujiang Hospital, Southern Medical University, No. 253, Industry Avenue, Haizhu District, Guangzhou, 510280, Guangdong, China
| | - Shufang Zhang
- Department of Gynecology, Southern University of Science and Technology Hospital, Shenzhen, 518000, China
| | - Dongxian Peng
- Obstetrics and Gynecology Center, Department of Gynecology, Zhujiang Hospital, Southern Medical University, No. 253, Industry Avenue, Haizhu District, Guangzhou, 510280, Guangdong, China.
| |
Collapse
|
40
|
Li M, Hao X, Hu Z, Tian J, Shi J, Ma D, Guo M, Li S, Zuo C, Liang Y, Tang M, Mao C, Xu Y, Shi C. Microvascular and cellular dysfunctions in Alzheimer's disease: an integrative analysis perspective. Sci Rep 2024; 14:20944. [PMID: 39251797 PMCID: PMC11385648 DOI: 10.1038/s41598-024-71888-0] [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: 05/08/2024] [Accepted: 09/02/2024] [Indexed: 09/11/2024] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia, characterized by memory loss, cognitive decline, personality changes, and various neurological symptoms. The role of blood-brain barrier (BBB) injury, extracellular matrix (ECM) abnormalities, and oligodendrocytes (ODCs) dysfunction in AD has gained increasing attention, yet the detailed pathogenesis remains elusive. This study integrates single-cell sequencing of AD patients' cerebrovascular system with a genome-wide association analysis. It aims to elucidate the associations and potential mechanisms behind pericytes injury, ECM disorder, and ODCs dysfunction in AD pathogenesis. Finally, we identified that abnormalities in the pericyte PI3K-AKT-FOXO signaling pathway may be involved in the pathogenic process of AD. This comprehensive approach sheds new light on the complex etiology of AD and opens avenues for advanced research into its pathogenesis and therapeutic strategies.
Collapse
Affiliation(s)
- Mengjie Li
- Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Xiaoyan Hao
- Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Zhengwei Hu
- Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Jie Tian
- Zhengzhou Railway Vocational and Technical College, Zhengzhou, 450000, Henan, China
| | - Jingjing Shi
- Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Dongrui Ma
- Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Mengnan Guo
- Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Shuangjie Li
- Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Chunyan Zuo
- Zhengzhou University, Zhengzhou, 450000, Henan, China
| | | | - Mibo Tang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, 1 Jian-she East Road, Zhengzhou, 450000, Henan, China
| | - Chengyuan Mao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, 1 Jian-she East Road, Zhengzhou, 450000, Henan, China
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, 1 Jian-she East Road, Zhengzhou, 450000, Henan, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, 450000, Henan, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Changhe Shi
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, 1 Jian-she East Road, Zhengzhou, 450000, Henan, China.
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China.
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, 450000, Henan, China.
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China.
| |
Collapse
|
41
|
Tian S, Zhong K, Yang Z, Fu J, Cai Y, Xiao M. Investigating the mechanism of tricyclic decyl benzoxazole -induced apoptosis in liver Cancer cells through p300-mediated FOXO3 activation. Cell Signal 2024; 121:111280. [PMID: 38960058 DOI: 10.1016/j.cellsig.2024.111280] [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: 04/10/2024] [Revised: 06/07/2024] [Accepted: 06/26/2024] [Indexed: 07/05/2024]
Abstract
OBJECTIVE To investigate whether tricyclic decylbenzoxazole (TDB) regulates liver cancer cell proliferation and apoptosis through p300-mediated FOXO acetylation. METHODS Sequencing, adenovirus, and lentivirus transfection were performed in human liver cancer cell line SMMC-7721 and apoptosis was detected by Tunel, Hoechst, and flow cytometry. TEM for mitochondrial morphology, MTT for cell proliferation ability, Western blot, and PCR were used to detect protein levels and mRNA changes. RESULTS Sequencing analysis and cell experiments confirmed that TDB can promote the up-regulation of FOXO3 expression. TDB induced FOXO3 up-regulation in a dose-dependent manner, promoted the expression of p300 and Bim, and enhanced the acetylation and dephosphorylation of FOXO3, thus promoting apoptosis. p300 promotes apoptosis of cancer cells through Bim and other proteins, while HAT enhances the phosphorylation of FOXO3 and inhibits apoptosis. Overexpression of FOXO3 can increase the expression of exo-apoptotic pathways (FasL, TRAIL), endo-apoptotic pathways (Bim), and acetylation at the protein level and inhibit cell proliferation and apoptotic ability, while FOXO3 silencing or p300 mutation can partially reverse apoptosis. In tumor tissues with overexpression of FOXO3, TDB intervention can further increase the expression of p53 and caspase-9 proteins in tumor cells, resulting in loss of mitochondrial membrane integrity during apoptosis, the release of cytoplasm during signal transduction, activation of caspase-9 and synergistic inhibition of growth. CONCLUSION TDB induces proliferation inhibition and promotes apoptosis of SMMC-7721 cells by activating p300-mediated FOXO3 acetylation.
Collapse
Affiliation(s)
- Shuhong Tian
- Research Center for Drug Safety Evaluation of Hainan Province, Hainan Medical University, Haikou 571199, China
| | - Keyan Zhong
- Clinical Skills Experimental Teaching Center of Hainan Medical University, Haikou 571199, China
| | - Zhaoxin Yang
- Research Center for Drug Safety Evaluation of Hainan Province, Hainan Medical University, Haikou 571199, China
| | - Jian Fu
- Research Center for Drug Safety Evaluation of Hainan Province, Hainan Medical University, Haikou 571199, China
| | - Yangbo Cai
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou 570100, China
| | - Min Xiao
- Research Center for Drug Safety Evaluation of Hainan Province, Hainan Medical University, Haikou 571199, China.
| |
Collapse
|
42
|
Wang P, Zou K, Cao J, Zhang Z, Yuan W, Chen J, Xu J, Zou Z, Chen D, Ruan H, Feng J, Lin X, Jin H. Protein phosphatase SCP4 regulates cartilage development and endochondral osteogenesis via FoxO3a dephosphorylation. Cell Prolif 2024; 57:e13691. [PMID: 38886174 PMCID: PMC11503251 DOI: 10.1111/cpr.13691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 05/02/2024] [Accepted: 05/31/2024] [Indexed: 06/20/2024] Open
Abstract
The regulatory mechanisms involved in embryonic development are complex and yet remain unclear. SCP4 represents a novel nucleus-resident phosphatase identified in our previous study. The primary aim of this study was to elucidate the function of SCP4 in the progress of cartilage development and endochondral osteogenesis. SCP4-/- and SCP4Col2ER mice were constructed to assess differences in bone formation using whole skeleton staining. ABH/OG staining was used to compare chondrocyte differentiation and cartilage development. Relevant biological functions were analysed using RNA-sequencing and GO enrichment, further validated by immunohistochemical staining, Co-IP and Western Blot. Global SCP4 knockout led to abnormal embryonic development in SCP4-/- mice, along with delayed endochondral osteogenesis. In parallel, chondrocyte-specific removal of SCP4 yielded more severe embryonic deformities in SCP4Col2ER mice, including limb shortening, reduced chondrocyte number in the growth plate, disorganisation and cell enlargement. Moreover, RNA-sequencing analysis showed an association between SCP4 and chondrocyte apoptosis. Notably, Tunnel-positive cells were indeed increased in the growth plates of SCP4Col2ER mice. The deficiency of SCP4 up-regulated the expression levels of pro-apoptotic proteins both in vivo and in vitro. Additionally, phosphorylation of FoxO3a (pFoxO3a), a substrate of SCP4, was heightened in chondrocytes of SCP4Col2ER mice growth plate, and the direct interaction between SCP4 and pFoxO3a was further validated in chondrocytes. Our findings underscore the critical role of SCP4 in regulating cartilage development and endochondral osteogenesis during embryonic development partially via inhibition of chondrocytes apoptosis regulated by FoxO3a dephosphorylation.
Collapse
Affiliation(s)
- Pinger Wang
- Institute of Orthopedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Provincial Hospital of Chinese MedicineHangzhouZhejiangChina
- The First College of Clinical MedicineZhejiang Chinese Medical UniversityHangzhouZhejiangChina
| | - Kaiao Zou
- Institute of Orthopedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Provincial Hospital of Chinese MedicineHangzhouZhejiangChina
- The First College of Clinical MedicineZhejiang Chinese Medical UniversityHangzhouZhejiangChina
| | - Jin Cao
- The MOE Key Laboratory of Biosystems Homeostasis and Protection and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences InstituteZhejiang UniversityHangzhouZhejiangChina
| | - Zhengmao Zhang
- Department of Pathology and Laboratory MedicineWeill Cornell MedicineNew York CityNew YorkUSA
| | - Wenhua Yuan
- Institute of Orthopedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Provincial Hospital of Chinese MedicineHangzhouZhejiangChina
- The First College of Clinical MedicineZhejiang Chinese Medical UniversityHangzhouZhejiangChina
| | - Jiali Chen
- Institute of Orthopedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Provincial Hospital of Chinese MedicineHangzhouZhejiangChina
- The First College of Clinical MedicineZhejiang Chinese Medical UniversityHangzhouZhejiangChina
| | - Jianbo Xu
- Institute of Orthopedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Provincial Hospital of Chinese MedicineHangzhouZhejiangChina
- The First College of Clinical MedicineZhejiang Chinese Medical UniversityHangzhouZhejiangChina
| | - Zhen Zou
- Institute of Orthopedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Provincial Hospital of Chinese MedicineHangzhouZhejiangChina
- The First College of Clinical MedicineZhejiang Chinese Medical UniversityHangzhouZhejiangChina
| | - Di Chen
- Research Center for Computer‐aided Drug DiscoveryChinese Academy of Sciences, Shenzhen Institute of Advanced TechnologyShenzhenGuangdongChina
- Faculty of Pharmaceutical SciencesChinese Academy of Sciences, Shenzhen Institute of Advanced TechnologyShenzhenGuangdongChina
| | - Hongfeng Ruan
- Institute of Orthopedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Provincial Hospital of Chinese MedicineHangzhouZhejiangChina
- The First College of Clinical MedicineZhejiang Chinese Medical UniversityHangzhouZhejiangChina
| | - Jianying Feng
- School of StomatologyZhejiang Chinese Medical UniversityHangzhouZhejiangChina
| | - Xia Lin
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
| | - Hongting Jin
- Institute of Orthopedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Provincial Hospital of Chinese MedicineHangzhouZhejiangChina
- The First College of Clinical MedicineZhejiang Chinese Medical UniversityHangzhouZhejiangChina
| |
Collapse
|
43
|
Niu Q, Lin C, Yang S, Rong S, Wei J, Zhao T, Peng Y, Cheng Z, Xie Y, Wang Y. FoxO1-Overexpressed Small Extracellular Vesicles Derived from hPDLSCs Promote Periodontal Tissue Regeneration by Reducing Mitochondrial Dysfunction to Regulate Osteogenesis and Inflammation. Int J Nanomedicine 2024; 19:8751-8768. [PMID: 39220194 PMCID: PMC11365494 DOI: 10.2147/ijn.s470419] [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: 03/24/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024] Open
Abstract
Purpose Periodontitis is a chronic infectious disease characterized by progressive inflammation and alveolar bone loss. Forkhead box O1 (FoxO1), an important regulator, plays a crucial role in maintaining bone homeostasis and regulating macrophage energy metabolism and osteogenic differentiation of mesenchymal stem cells (MSCs). In this study, FoxO1 was overexpressed into small extracellular vesicles (sEV) using engineering technology, and effects of FoxO1-overexpressed sEV on periodontal tissue regeneration as well as the underlying mechanisms were investigated. Methods Human periodontal ligament stem cell (hPDLSCs)-derived sEV (hPDLSCs-sEV) were isolated using ultracentrifugation. They were then characterized using transmission electron microscopy, Nanosight, and Western blotting analyses. hPDLSCs were treated with hPDLSCs-sEV in vitro after stimulation with lipopolysaccharide, and osteogenesis was evaluated. The effect of hPDLSCs-sEV on the polarization phenotype of THP-1 macrophages was also evaluated. In addition, we measured the reactive oxygen species (ROS) levels, adenosine triphosphate (ATP) production, mitochondrial characteristics, and metabolism of hPDLSCs and THP-1 cells. Experimental periodontitis was established in vivo in mice. HPDLSCs-sEV or phosphate-buffered saline (PBS) were injected into periodontal tissues for four weeks, and the maxillae were collected and assessed by micro-computed tomography, histological staining, and small animal in vivo imaging. Results In vitro, FoxO1-overexpressed sEV promoted osteogenic differentiation of hPDLSCs in the inflammatory environment and polarized THP-1 cells from the M1 phenotype to the M2 phenotype. Furthermore, FoxO1-overexpressed sEV regulated the ROS level, ATP production, mitochondrial characteristics, and metabolism of hPDLSCs and THP-1 cells in the inflammatory environment. In the in vivo analyses, FoxO1-overexpressed sEV effectively promoted bone formation and inhibited inflammation. Conclusion FoxO1-overexpressed sEV can regulate osteogenesis and immunomodulation. The ability of FoxO1-overexpressed sEV to regulate inflammation and osteogenesis can pave the way for the establishment of a therapeutic approach for periodontitis.
Collapse
Affiliation(s)
- Qingru Niu
- Hospital of Stomatology, Guanghua School of Stomatology, Guangzhou, People’s Republic of China
- Sun Yat-Sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| | - Chuanmiao Lin
- Hospital of Stomatology, Guanghua School of Stomatology, Guangzhou, People’s Republic of China
- Sun Yat-Sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| | - Shuqing Yang
- Hospital of Stomatology, Guanghua School of Stomatology, Guangzhou, People’s Republic of China
- Sun Yat-Sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| | - Shuxuan Rong
- Hospital of Stomatology, Guanghua School of Stomatology, Guangzhou, People’s Republic of China
- Sun Yat-Sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| | - Junbin Wei
- Hospital of Stomatology, Guanghua School of Stomatology, Guangzhou, People’s Republic of China
- Sun Yat-Sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| | - Tingting Zhao
- Hospital of Stomatology, Guanghua School of Stomatology, Guangzhou, People’s Republic of China
- Sun Yat-Sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| | - Yingying Peng
- Hospital of Stomatology, Guanghua School of Stomatology, Guangzhou, People’s Republic of China
- Sun Yat-Sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| | - Zhilan Cheng
- Hospital of Stomatology, Guanghua School of Stomatology, Guangzhou, People’s Republic of China
- Sun Yat-Sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| | - Yunyi Xie
- Hospital of Stomatology, Guanghua School of Stomatology, Guangzhou, People’s Republic of China
- Sun Yat-Sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| | - Yan Wang
- Hospital of Stomatology, Guanghua School of Stomatology, Guangzhou, People’s Republic of China
- Sun Yat-Sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| |
Collapse
|
44
|
Xu P, Wang J, Wang J, Hu X, Wang W, Lu S, Sheng Y. Anoectochilus roxburghii Extract Extends the Lifespan of Caenorhabditis elegans through Activating the daf-16/FoxO Pathway. Antioxidants (Basel) 2024; 13:945. [PMID: 39199191 PMCID: PMC11351832 DOI: 10.3390/antiox13080945] [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: 07/12/2024] [Revised: 07/25/2024] [Accepted: 07/30/2024] [Indexed: 09/01/2024] Open
Abstract
As a significant global issue, aging is prompting people's interest in the potential anti-aging properties of Anoectochilus roxburghii (A. roxburghii), a plant traditionally utilized in various Asian countries for its purported benefits in treating diabetes and combating aging. However, the specific anti-aging components and mechanisms of A. roxburghii remain unclear. This study aims to investigate the anti-aging effects and mechanisms of A. roxburghii extract E (ARE). Caenorhabditis elegans (C. elegans) were exposed to media containing different concentrations of ARE whose superior in vitro radical scavenging capacity was thus identified. Lifespan assays, stress resistance tests, and RT-qPCR analyses were conducted to evaluate anti-aging efficacy, reactive oxygen species (ROS) levels, antioxidant enzyme activity, and daf-16, sod-3, and gst-4 levels. Additionally, transcriptomic and metabolomic analyses were performed to elucidate the potential anti-aging mechanisms of ARE. Fluorescence protein assays and gene knockout experiments were employed to validate the impacts of ARE on anti-aging mechanisms. Our results revealed that ARE not only prolonged the lifespan of C. elegans but also mitigated ROS and lipofuscin accumulation, and boosted resistance to UV and heat stress. Furthermore, ARE modulated the expression of pivotal anti-aging genes including daf-16, sod-3, and gst-4, facilitating the nuclear translocation of DAF-16. Significantly, ARE failed to extend the lifespan of daf-16-deficient C. elegans (CF1038), indicating its dependency on the daf-16/FoxO signaling pathway. These results underscored the effectiveness of ARE as a natural agent for enhancing longevity and stress resilience to C. elegans, potentially to human.
Collapse
Affiliation(s)
- Peng Xu
- Xingzhi College, Zhejiang Normal University, Jinhua 321100, China; (P.X.); (J.W.); (X.H.)
- School of Basic Medical Science, Hangzhou Normal University, Hangzhou 311121, China
| | - Jianfeng Wang
- Xingzhi College, Zhejiang Normal University, Jinhua 321100, China; (P.X.); (J.W.); (X.H.)
| | - Junyi Wang
- Life Sciences, Zhejiang Normal University, Jinhua 321017, China;
| | - Xiaoxiao Hu
- Xingzhi College, Zhejiang Normal University, Jinhua 321100, China; (P.X.); (J.W.); (X.H.)
| | - Wei Wang
- Taizhou Research Institute, Southern University of Science and Technology, Taizhou 317700, China;
| | - Shengmin Lu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Food Science, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Yingkun Sheng
- Xingzhi College, Zhejiang Normal University, Jinhua 321100, China; (P.X.); (J.W.); (X.H.)
| |
Collapse
|
45
|
Zhang H, Bai Y, Li J, Chen T, Shang G. FBXO22 promotes osteosarcoma progression via regulation of FOXO1 for ubiquitination and degradation. J Cell Mol Med 2024; 28:e70021. [PMID: 39153212 PMCID: PMC11330286 DOI: 10.1111/jcmm.70021] [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: 04/23/2024] [Revised: 07/26/2024] [Accepted: 08/07/2024] [Indexed: 08/19/2024] Open
Abstract
Accumulating evidence has demonstrated that F-box protein 22 (FBXO22) participates in tumour development and progression in various types of human malignancies. However, the functions and detailed molecular mechanisms of FBXO22 in osteosarcoma tumorigenesis and progression remain elusive. In this study, we aimed to determine the effects of FBXO22 on the cell proliferation, migration and invasion of osteosarcoma cells using cell counting kit-8 and Matrigel Transwell approaches. Moreover, we explored the molecular mechanisms by which FBXO22 mediated oncogenesis and progression in osteosarcoma via Western blotting, immunoprecipitation and ubiquitination. We found that FBXO22 depletion inhibited the proliferation, migration and invasion of osteosarcoma cells, whereas FBXO22 overexpression increased the proliferation and motility of osteosarcoma cells. Mechanistically, FBXO22 promoted the ubiquitination and degradation of FoxO1 in osteosarcoma cells. FBXO22 depletion reduced cell proliferation and motility via regulation of FoxO1. Taken together, our findings provide new insight into FBXO22-induced osteosarcoma tumorigenesis. The inhibition of FBXO22 could be a promising strategy for the treatment of osteosarcoma.
Collapse
Affiliation(s)
- He Zhang
- Department of OrthopedicsShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Yang Bai
- Department of NursingShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Jiatong Li
- Department of OrthopedicsShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Ting Chen
- Department of OrthopedicsShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Guanning Shang
- Department of OrthopedicsShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| |
Collapse
|
46
|
Kim HY, Jang HJ, Muthamil S, Shin UC, Lyu JH, Kim SW, Go Y, Park SH, Lee HG, Park JH. Novel insights into regulators and functional modulators of adipogenesis. Biomed Pharmacother 2024; 177:117073. [PMID: 38981239 DOI: 10.1016/j.biopha.2024.117073] [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: 04/15/2024] [Revised: 06/27/2024] [Accepted: 06/29/2024] [Indexed: 07/11/2024] Open
Abstract
Adipogenesis is a process that differentiates new adipocytes from precursor cells and is tightly regulated by several factors, including many transcription factors and various post-translational modifications. Recently, new roles of adipogenesis have been suggested in various diseases. However, the molecular mechanisms and functional modulation of these adipogenic genes remain poorly understood. This review summarizes the regulatory factors and modulators of adipogenesis and discusses future research directions to identify novel mechanisms regulating adipogenesis and the effects of adipogenic regulators in pathological conditions. The master adipogenic transcriptional factors PPARγ and C/EBPα were identified along with other crucial regulatory factors such as SREBP, Kroxs, STAT5, Wnt, FOXO1, SWI/SNF, KLFs, and PARPs. These transcriptional factors regulate adipogenesis through specific mechanisms, depending on the adipogenic stage. However, further studies related to the in vivo role of newly discovered adipogenic regulators and their function in various diseases are needed to develop new potent therapeutic strategies for metabolic diseases and cancer.
Collapse
Affiliation(s)
- Hyun-Yong Kim
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do 58245, Republic of Korea; New Drug Development Center, Osong Medical Innovation Foundation, 123, Osongsaengmyeong-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28160, Republic of Korea.
| | - Hyun-Jun Jang
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do 58245, Republic of Korea; Research Group of Personalized Diet, Korea Food Research Institute, Wanju-gun, Jeollabuk-do 55365, Republic of Korea.
| | - Subramanian Muthamil
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do 58245, Republic of Korea.
| | - Ung Cheol Shin
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do 58245, Republic of Korea.
| | - Ji-Hyo Lyu
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do 58245, Republic of Korea.
| | - Seon-Wook Kim
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do 58245, Republic of Korea.
| | - Younghoon Go
- Korean Medicine (KM)-application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea.
| | - Seong-Hoon Park
- Genetic and Epigenetic Toxicology Research Group, Korea Institute of Toxicology, Daejeon 34141, Republic of Korea.
| | - Hee Gu Lee
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea.
| | - Jun Hong Park
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do 58245, Republic of Korea; University of Science & Technology (UST), KIOM campus, Korean Convergence Medicine Major, Daejeon 34054, Republic of Korea.
| |
Collapse
|
47
|
Yadav P, Singh SK, Datta S, Verma S, Verma A, Rakshit A, Bali A, Bhatti JS, Khurana A, Navik U. Therapeutic potential and pharmacological mechanism of visnagin. JOURNAL OF INTEGRATIVE MEDICINE 2024; 22:399-412. [PMID: 38797603 DOI: 10.1016/j.joim.2024.05.001] [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: 05/18/2023] [Accepted: 04/10/2024] [Indexed: 05/29/2024]
Abstract
Visnagin is a furanochromone and one of the most important compound in the Ammi visnaga (L.) Lam (a synonym of Visnaga daucoides Gaertn.) plant, which is used to cure various ailments. Many investigations into the bioactive properties of visnagin have been studied to date. The literature on visnagin demonstrates its biological properties, including anti-inflammatory, anti-diabetic, and beneficial effects in cardiovascular and renal diseases. Moreover, visnagin improves sperm quality parameters, stimulates steroidogenesis, and increases serum gonadotropins and testosterone levels, while decreasing pro-inflammatory cytokines, oxidative damage, genomic instability, and it modulates apoptosis. Thus, visnagin has emerged as an exciting lead for further research, owing to its potential in various unmet clinical needs. The current review summarized its basic structure, pharmacokinetics, and pharmacological effects, focusing on its mechanisms of action. The review will help to understand the potential of visnagin as an alternative treatment strategy for several diseases and provide insight into research topics that need further exploration for visnagin's safe clinical use. Please cite this article as: Yadav P, Singh SK, Datta S, Verma S, Verma A, Rakshit A, Bali A, Bhatti JS, Khurana A, Navik U. Therapeutic potential and pharmacological mechanism of visnagin. J Integr Med. 2024; 22(4): 399-412.
Collapse
Affiliation(s)
- Poonam Yadav
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Sumeet Kumar Singh
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Sayantap Datta
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204-5000, USA
| | - Saloni Verma
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Aarti Verma
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Arnab Rakshit
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Anjana Bali
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Jasvinder Singh Bhatti
- Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India
| | - Amit Khurana
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India.
| | - Umashanker Navik
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab 151401, India.
| |
Collapse
|
48
|
Peng ZQ, Guan XH, Yu ZP, Wu J, Han XH, Li MH, Qu XH, Chen ZP, Han XJ, Wang XY. Human amniotic mesenchymal stem cells-derived conditioned medium and exosomes alleviate oxidative stress-induced retinal degeneration by activating PI3K/Akt/FoxO3 pathway. Exp Eye Res 2024; 244:109919. [PMID: 38729254 DOI: 10.1016/j.exer.2024.109919] [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: 02/29/2024] [Revised: 04/01/2024] [Accepted: 04/13/2024] [Indexed: 05/12/2024]
Abstract
Age-related macular degeneration (AMD) is the leading cause of vision loss among the elderly, which is primarily attributed to oxidative stress-induced damage to the retinal pigment epithelium (RPE). Human amniotic mesenchymal stem cells (hAMSC) were considered to be one of the most promising stem cells for clinical application due to their low immunogenicity, tissue repair ability, pluripotent potential and potent paracrine effects. The conditional medium (hAMSC-CM) and exosomes (hAMSC-exo) derived from hAMSC, as mediators of intercellular communication, play an important role in the treatment of retinal diseases, but their effect and mechanism on oxidative stress-induced retinal degeneration are not explored. Here, we reported that hAMSC-CM alleviated H2O2-induced ARPE-19 cell death through inhibiting mitochondrial-mediated apoptosis pathway in vitro. The overproduction of reactive oxygen species (ROS), alteration in mitochondrial morphology, loss of mitochondrial membrane potential and elevation of Bax/Bcl2 ratio in ARPE-19 cells under oxidative stress were efficiently reversed by hAMSC-CM. Moreover, it was found that hAMSC-CM protected cells against oxidative injury via PI3K/Akt/FoxO3 signaling. Intriguingly, exosome inhibitor GW4869 alleviated the inhibitory effect of hAMSC-CM on H2O2-induced decrease in cell viability of ARPE-19 cells. We further demonstrated that hAMSC-exo exerted the similar protective effect on ARPE-19 cells against oxidative damage as hAMSC-CM. Additionally, both hAMSC-CM and hAMSC-exo ameliorated sodium iodate-induced deterioration of RPE and retinal damage in vivo. These results first indicate that hAMSC-CM and hAMSC-exo protect RPE cells from oxidative damage by regulating PI3K/Akt/FoxO3 pathway, suggesting hAMSC-CM and hAMSC-exo will be a promising cell-free therapy for the treatment of AMD in the future.
Collapse
Affiliation(s)
- Zhe-Qing Peng
- Institute of Geriatrics, Jiangxi Provincial People's Hospital & the First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330006, PR China; Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Xiao-Hui Guan
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330031, PR China
| | - Zhen-Ping Yu
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330031, PR China
| | - Jie Wu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, 330052, PR China
| | - Xin-Hao Han
- Institute of Geriatrics, Jiangxi Provincial People's Hospital & the First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330006, PR China
| | - Ming-Hui Li
- Institute of Geriatrics, Jiangxi Provincial People's Hospital & the First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330006, PR China; Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Xin-Hui Qu
- Institute of Geriatrics, Jiangxi Provincial People's Hospital & the First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330006, PR China; The Second Department of Neurology, Jiangxi Provincial People's Hospital & the First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330006, PR China
| | - Zhi-Ping Chen
- Institute of Geriatrics, Jiangxi Provincial People's Hospital & the First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330006, PR China
| | - Xiao-Jian Han
- Institute of Geriatrics, Jiangxi Provincial People's Hospital & the First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330006, PR China.
| | - Xiao-Yu Wang
- Institute of Geriatrics, Jiangxi Provincial People's Hospital & the First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, 330006, PR China.
| |
Collapse
|
49
|
Jiang C, Yao D, Liu Z, Zheng Y, Chen M, Yim WY, Zheng Q, Zhang T, Fan L, Fan Z, Geng B, Tian R, Zhou T, Qiao W, Shi J, Li F, Xu L, Huang Y, Dong N. FOXO1 regulates RUNX2 ubiquitination through SMURF2 in calcific aortic valve disease. Redox Biol 2024; 73:103215. [PMID: 38810422 PMCID: PMC11167395 DOI: 10.1016/j.redox.2024.103215] [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: 04/19/2024] [Revised: 05/16/2024] [Accepted: 05/24/2024] [Indexed: 05/31/2024] Open
Abstract
The prevalence of calcific aortic valve disease (CAVD) remains substantial while there is currently no medical therapy available. Forkhead box O1 (FOXO1) is known to be involved in the pathogenesis of cardiovascular diseases, including vascular calcification and atherosclerosis; however, its specific role in calcific aortic valve disease remains to be elucidated. In this study, we identified FOXO1 significantly down-regulated in the aortic valve interstitial cells (VICs) of calcified aortic valves by investigating clinical specimens and GEO database analysis. FOXO1 silencing or inhibition promoted VICs osteogenic differentiation in vitro and aortic valve calcification in Apoe-/- mice, respectively. We identified that FOXO1 facilitated the ubiquitination and degradation of RUNX2, which process was mainly mediated by SMAD-specific E3 ubiquitin ligase 2 (SMURF2). Our discoveries unveil a heretofore unacknowledged mechanism involving the FOXO1/SMURF2/RUNX2 axis in CAVD, thereby proposing the potential therapeutic utility of FOXO1 or SMURF2 as viable strategies to impede the progression of CAVD.
Collapse
Affiliation(s)
- Chen Jiang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Dingyi Yao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Zongtao Liu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Yidan Zheng
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Ming Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Wai Yen Yim
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Qiang Zheng
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Tailong Zhang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Lin Fan
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Zhengfeng Fan
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Bingchuan Geng
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Rui Tian
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Tingwen Zhou
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Weihua Qiao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Jiawei Shi
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Fei Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China.
| | - Li Xu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China.
| | - Yuming Huang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China.
| |
Collapse
|
50
|
Ebrahimnezhad M, Valizadeh A, Majidinia M, Tabnak P, Yousefi B. Unveiling the potential of FOXO3 in lung cancer: From molecular insights to therapeutic prospects. Biomed Pharmacother 2024; 176:116833. [PMID: 38843589 DOI: 10.1016/j.biopha.2024.116833] [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: 02/24/2024] [Revised: 05/18/2024] [Accepted: 05/26/2024] [Indexed: 06/20/2024] Open
Abstract
Lung cancer poses a significant challenge regarding molecular heterogeneity, as it encompasses a wide range of molecular alterations and cancer-related pathways. Recent discoveries made it feasible to thoroughly investigate the molecular mechanisms underlying lung cancer, giving rise to the possibility of novel therapeutic strategies relying on molecularly targeted drugs. In this context, forkhead box O3 (FOXO3), a member of forkhead transcription factors, has emerged as a crucial protein commonly dysregulated in cancer cells. The regulation of the FOXO3 in reacting to external stimuli plays a key role in maintaining cellular homeostasis as a component of the molecular machinery that determines whether cells will survive or dies. Indeed, various extrinsic cues regulate FOXO3, affecting its subcellular location and transcriptional activity. These regulations are mediated by diverse signaling pathways, non-coding RNAs (ncRNAs), and protein interactions that eventually drive post-transcriptional modification of FOXO3. Nevertheless, while it is no doubt that FOXO3 is implicated in numerous aspects of lung cancer, it is unclear whether they act as tumor suppressors, promotors, or both based on the situation. However, FOXO3 serves as an intriguing possible target in lung cancer therapeutics while widely used anti-cancer chemo drugs can regulate it. In this review, we describe a summary of recent findings on molecular mechanisms of FOXO3 to clarify that targeting its activity might hold promise in lung cancer treatment.
Collapse
Affiliation(s)
- Mohammad Ebrahimnezhad
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Amir Valizadeh
- Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Majidinia
- Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Peyman Tabnak
- Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Bahman Yousefi
- Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|