51
|
Deng J, Qin J, Song G, Li C, Tang W, Tang Y, Xiao X, Wu L, He S, Zhou Y, Li J, Wang Y. The potential of low-intensity pulsed ultrasound to apply the long-term ovary protection from injury induced by 4-vinylcyclohexene diepoxide through inhibiting granulosa cell apoptosis. Bioeng Transl Med 2025; 10:e10744. [PMID: 40385545 PMCID: PMC12079353 DOI: 10.1002/btm2.10744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 12/07/2024] [Accepted: 12/07/2024] [Indexed: 01/04/2025] Open
Abstract
The potential of low-intensity pulsed ultrasound (LIPUS) in regulating ovarian function has been demonstrated; however, there is a lack of scientific evidence regarding the long-term efficacy of LIPUS in treating ovarian injury and understanding its regulatory mechanisms. In this study, 4-vinylcyclohexene diepoxide (VCD) was used to induce ovarian injury in rats, and LIPUS was applied to target the damaged ovarian tissues. The research aimed to investigate the long-term protective effect of LIPUS against ovum toxicity induced by VCD and elucidate the associated molecular mechanisms. During the experiment, HE staining was employed for observing the morphology and structure of the ovary, while protein sequencing was utilized for identifying and confirming the molecular mechanism through which LIPUS restores the damaged ovarian structure. The long-term effectiveness of LIPUS in protecting against ovarian injury was evaluated through ELISA, estrous cycle monitoring, fertility testing, and behavioral analysis. The results indicated that LIPUS effectively restored the structure of damaged ovaries. Both in vivo and in vitro studies revealed that this protective effect may be attributed to LIPUS inhibiting apoptosis of ovarian granulosa cells (GCs) by regulating Daxx-mediated ASK1/JNK signaling pathway. Subsequent functional tests demonstrated significant improvements in sex hormone secretion and regulation of estrous cycle within 6 cycles following LIPUS treatment. Additionally, there was a notable increase in offspring numbers after mating. Behavioral analysis revealed that LIPUS effectively alleviated menopausal symptoms resulting from ovarian injury including mood fluctuations, cognitive behavior changes, and reduced muscle excitability levels. These findings suggest that beneficial effects of LIPUS may help reduce VCD-induced ovarian damage with long-term efficacy.
Collapse
Affiliation(s)
- Juan Deng
- State Key Laboratory of Ultrasound in Medicine and EngineeringChongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Biomedical EngineeringChongqing Medical UniversityChongqingChina
| | - Juan Qin
- Department of Obstetrics and Gynecology, Guiyang Maternal and Child Health Care HospitalGuizhou Medical UniversityGuizhouChina
| | - Guolin Song
- Department of EmergencyThe Second Affiliated Hospital of Guizhou University of Traditional Chinese MedicineGuizhouChina
| | - Chenghai Li
- State Key Laboratory of Ultrasound in Medicine and EngineeringChongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Biomedical EngineeringChongqing Medical UniversityChongqingChina
| | - Wentao Tang
- State Key Laboratory of Ultrasound in Medicine and EngineeringChongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Biomedical EngineeringChongqing Medical UniversityChongqingChina
| | - Yilin Tang
- State Key Laboratory of Ultrasound in Medicine and EngineeringChongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Biomedical EngineeringChongqing Medical UniversityChongqingChina
| | - Xinfang Xiao
- State Key Laboratory of Ultrasound in Medicine and EngineeringChongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Biomedical EngineeringChongqing Medical UniversityChongqingChina
| | - Liu Wu
- State Key Laboratory of Ultrasound in Medicine and EngineeringChongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Biomedical EngineeringChongqing Medical UniversityChongqingChina
| | - Sicheng He
- State Key Laboratory of Ultrasound in Medicine and EngineeringChongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Biomedical EngineeringChongqing Medical UniversityChongqingChina
| | - Yiqing Zhou
- State Key Laboratory of Ultrasound in Medicine and EngineeringChongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Biomedical EngineeringChongqing Medical UniversityChongqingChina
| | - Junfen Li
- State Key Laboratory of Ultrasound in Medicine and EngineeringChongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Biomedical EngineeringChongqing Medical UniversityChongqingChina
| | - Yan Wang
- State Key Laboratory of Ultrasound in Medicine and EngineeringChongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Biomedical EngineeringChongqing Medical UniversityChongqingChina
| |
Collapse
|
52
|
Montazeri V, Varshosaz P, Fakhrjou A, Pirouzpanah S. Hormone Receptor-Dependent Correlations Between Angiopoietins and VEGF-C in Primary Breast Cancer: Insights Into Lymphangiogenic Biomarkers. Cancer Rep (Hoboken) 2025; 8:e70101. [PMID: 40348609 PMCID: PMC12063722 DOI: 10.1002/cnr2.70101] [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: 11/21/2023] [Revised: 12/02/2024] [Accepted: 12/10/2024] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND Biomarkers of angiogenesis and lymphangiogenesis have been explored in cancer prognostic models; however, their potential role in assessing local tumor invasiveness remains poorly understood. AIMS This study aimed to evaluate the correlations of angiogenic biomarkers, specifically the angiopoietin (ANG)-Tie system and vascular endothelial growth factor-C (VEGF-C), with lymphangiogenesis and the related histopathological characteristics in Iranian women with breast cancer. METHODS AND RESULTS In this consecutive case series (n = 149) from the Breast Cancer Risk and Lifestyle (BCRL) study, plasma levels of pro-angiogenic factors, including VEGF-C, ANGs, and Tie-2, were assessed using ELISA. Clinicopathological data were collected, excluding stage IV cases to focus on patients with localized disease. Axillary lymph node metastasis (ANLM), and vascular invasion (VI) were common in the study population, occurring in 61.5% and 77.6% of cases, respectively (p < 0.01). Estrogen receptor-positive (ER+) tumors were observed in 89.1% of ANLM+ participants, while human epidermal growth factor receptor-2-positive (HER-2+) tumors were identified in 22.8% of patients with ALNM. Plasma levels of ANG-1 (r = 0.19) and VEGF-C (r = 0.29) were positively correlated with the ALNM ratio (p < 0.05). Multivariate analysis in patients with grade II tumors revealed significant inverse correlations between VEGF-C and angiogenic biomarkers, including ANG-2 (β = -0.25), the ANG-2/Tie-2 ratio (β = -0.28), and the (ANG-1 + ANG-2)/Tie-2 ratio (β = -0.29) (p < 0.05). Receiver operating characteristic (ROC) curve analysis indicated that ANG-2 could effectively assess ALNM status, with an optimal cutoff of 3.39 pg/mL, identifying ALNM in 66.0% of patients with low VEGF-C levels (95% CI: 0.54-0.78), increasing to 68.0% when combined with ANG-1 as the ANGs/Tie-2 ratio (95% CI: 0.56-0.80). In ER+ tumors, high plasma ANG-2 levels were observed (p < 0.05). Significantly higher levels of the (ANG-1 + ANG-2)/VEGF-C ratio were noted in patients with VI+ (p < 0.05). Findings descriptively highlighted ER+ status as a common characteristic in VI+ and ALNM+ tumors. In HER-2+ patients, both ANG-1 and the (ANG-1 + ANG-2)/Tie-2 ratio showed inverse correlations with VEGF-C, while in ER- breast cancer patients, ANG-2 was inversely correlated with VEGF-C. CONCLUSION These findings provide new insights into the inverse correlation between plasma levels of ANGs and VEGF-C, particularly in cases with positive ALNM, underscoring the role of hormone receptor-dependent characteristics. The integration of the triple angiogenic biomarkers ANG-2/Tie-2/VEGF-C within the tumor microenvironment, combined with the regulatory influence of hormonal receptors, merits further investigation as a potential biomarker panel for identifying lymphatic anomalies and VI positivity in breast cancer patients.
Collapse
Affiliation(s)
- Vahid Montazeri
- Drug Applied Research CenterTabriz University of Medical SciencesTabrizIran
- Molecular Medicine Research CenterTabriz University of Medical SciencesTabrizIran
- Department of Thoracic Surgery, Faculty of MedicineTabriz University of Medical SciencesTabrizIran
| | - Parisa Varshosaz
- Molecular Medicine Research CenterTabriz University of Medical SciencesTabrizIran
| | | | - Saeed Pirouzpanah
- Molecular Medicine Research CenterTabriz University of Medical SciencesTabrizIran
| |
Collapse
|
53
|
Xiang X, Huang L, Luo W, Qin L, Bian M, Chen W, Han G, Wang N, Mo G, Zhang C, Zhang Y, Yang H, Lu S, Zhang J, Fu T. Neuromuscular electrical stimulation alleviates stroke-related sarcopenia by promoting satellite cells myogenic differentiation via AMPK-ULK1-Autophagy axis. J Orthop Translat 2025; 52:249-264. [PMID: 40342549 PMCID: PMC12059223 DOI: 10.1016/j.jot.2025.03.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 03/11/2025] [Accepted: 03/31/2025] [Indexed: 05/11/2025] Open
Abstract
Background Stroke-related sarcopenia can result in muscle mass loss and muscle fibers abnormality, significantly affecting muscle function. The clinical management of stroke-related sarcopenia still requires further research and investigation. This study aims to explore a promising therapy to restore muscle function and promote muscle regeneration in stroke-related sarcopenia, providing a new theory for stroke-related sarcopenia treatment. Methods Stroke-related sarcopenia rat model was established by using permanent middle cerebral artery occlusion (pMCAO) rat and treated with neuromuscular electrical stimulation (NMES). Electrical stimulation (ES) treatment in vitro was mimicked to test the effect of NMES on muscle regeneration in rat skeletal muscle satellite cells (MuSCs). Catwalk, H&E and Masson's trichrome staining, immunofluorescence, transcriptomic analysis, transmission electron microscopy, MuSCs transfection, autophagy flux detection, quantitative real-time PCR analysis, Co-Immunoprecipitation and Western Blot were used to investigate the role of NMES and its mechanism in stroke-related sarcopenia in vivo. Results After NMES treatment, muscle mass and myogenic differentiation were significantly increased in stroke-related sarcopenia rats. The NMES group had more stable gait, neater footprints, higher muscle wet weight, more voluminous morphology and more regenerated muscle fibers. Additionally, ES treatment induced myogenic differentiation in rat MuSCs in vitro. Transcriptomic analysis also showed that "AMPK signaling pathway" was enriched and genes upregulated in ES-treated cells, revealing ES treatment could activate the autophagy in an AMPK-ULK1-dependent mechanism in MuSCs. Besides, it was also founded that infusion of AMPK or ULK1 inhibitor, knockdown of AMPK or ULK1 in MuSCs could block the effect of myotube formation of ES. Conclusion NMES not only restores muscle function but also enhances myogenic activity and muscle regeneration via AMPK-ULK1 autophagy in stroke-related sarcopenia rats. Our study provides a promising strategy for the treatment of stroke-related sarcopenia. The translational potential of this article This study first demonstrates that NMES alleviates stroke-related sarcopenia by promoting MuSCs differentiation through AMPK-ULK1-autophagy axis. The findings reveal a novel therapeutic mechanism, suggesting that NMES can restore muscle function and enhance regeneration in stroke patients. By combining NMES with MuSCs-based therapies, this approach offers a promising strategy for clinical rehabilitation, potentially improving muscle mass and function in stroke survivors. The translational potential lies in its applicability to non-invasive, cost-effective treatments for sarcopenia, enhancing patients' quality of life.
Collapse
Affiliation(s)
- Xingdong Xiang
- Department of Rehabilitation Medicine, Zhongshan Hospital Fudan University, Shanghai, 200032, China
| | - Lei Huang
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Wenchen Luo
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai, 200032, China
| | - Lieyang Qin
- Department of Cardiovascular Surgery, Zhongshan Hospital Fudan University, Shanghai, 200032, China
| | - Mengxuan Bian
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Weisin Chen
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Guanjie Han
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Ning Wang
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Guokang Mo
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Cheng Zhang
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Yongxing Zhang
- Department of Orthopedic Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Huilin Yang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215123, China
| | - Shunyi Lu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215123, China
| | - Jian Zhang
- Department of Rehabilitation Medicine, Zhongshan Hospital Fudan University, Shanghai, 200032, China
| | - Tengfei Fu
- Department of Rehabilitation Medicine, Zhongshan Hospital Fudan University, Shanghai, 200032, China
| |
Collapse
|
54
|
Han C, Fu S, Tang D, Chen Y, Liu D, Feng Z, Gou Y, Zhang C, Zhang W, Xiao L, Zhang J, Yi C, Xue Y, Peng D. Omic AI reveals new autophagy regulators from the Atg1 interactome in Saccharomyces cerevisiae. Front Cell Dev Biol 2025; 13:1554958. [PMID: 40365021 PMCID: PMC12069372 DOI: 10.3389/fcell.2025.1554958] [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: 01/03/2025] [Accepted: 04/16/2025] [Indexed: 05/15/2025] Open
Abstract
In Saccharomyces cerevisiae, Atg1 is a core autophagy-related (Atg) protein kinase (PK) in regulating macroautophagy/autophagy, by physically interacting with numerous other proteins, or by phosphorylating various substrates. It is unclear how many Atg1-interacting partners and substrates are also involved in regulating autophagy. Here, we conducted transcriptomic, proteomic and phosphoproteomic profiling of Atg1-dependent molecular landscapes during nitrogen starvation-triggered autophagy, and detected 244, 245 and 217 genes to be affected by ATG1 in the autophagic process at mRNA, protein, and phosphorylation levels, respectively. Based on the Atg1 interactome, we developed a novel artificial intelligence (AI) framework, inference of autophagy regulators from multi-omic data (iAMD), and predicted 12 Atg1-interacting partners and 17 substrates to be potentially functional in autophagy. Further experiments validated that Rgd1 and Whi5 are required for bulk autophagy, as well as physical interactions and co-localizations with Atg1 during autophagy. In particular, we demonstrated that 2 phosphorylation sites (p-sites), pS78 and pS149 of Whi5, are phosphorylated by Atg1 to regulate the formation of Atg1 puncta during autophagy initiation. A working model was illustrated to emphasize the importance of the Atg1-centered network in yeast autophagy. In addition, iAMD was extended to accurately predict Atg proteins and autophagy regulators from other PK interactomes, indicating a high transferability of the method. Taken together, we not only revealed new autophagy regulators from the Atg1 interactome, but also provided a useful resource for further analysis of yeast autophagy.
Collapse
Affiliation(s)
- Cheng Han
- MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shanshan Fu
- MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Dachao Tang
- MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuting Chen
- Department of Biochemistry, and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dan Liu
- MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zihao Feng
- MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yujie Gou
- MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chi Zhang
- MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Weizhi Zhang
- MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Leming Xiao
- MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jiayi Zhang
- MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Cong Yi
- Department of Biochemistry, and Department of Hepatobiliary and Pancreatic Surgery of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Xue
- MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Nanjing University Institute of Artificial Intelligence Biomedicine, Nanjing, Jiangsu, China
| | - Di Peng
- MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
55
|
Schein CH. Sequences and Structures of Viral Proteins Linked to the Genomes (VPg) of RNA Viruses. Viruses 2025; 17:645. [PMID: 40431656 PMCID: PMC12115650 DOI: 10.3390/v17050645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/23/2025] [Accepted: 04/27/2025] [Indexed: 05/29/2025] Open
Abstract
In the mid-1970s, it was revealed that the 5' end of the RNA genome of poliovirus (PV) was covalently linked to a peptide called VPg (viral protein, genome-linked). Subsequently, VPgs have been found attached to many other viruses and even phages. This review summarizes the patterns of physicochemical properties that are conserved within the VPgs of plus-strand RNA viruses where short-peptide VPgs have been identified. Mutagenesis and structural data indicate the importance of a 5 aa conserved motif at the N-termini of picornaviral VPgs (around the tyrosine 3 residue, which forms a covalent bond to UMP and the RNA). Hidden Markov models have been used to find motifs and VPgs in additional genera of picornaviruses, as well as dicistroviruses in insects and comoviruses in plants. These latter VPgs are bound to the RNA termina through linkages to serine or threonine. The role of free VPg and VPgpU needs clarification, especially in light of multiple genome copies in many of the viruses. Lysine and other positively charged side chains are hallmarks of VPgs. These may contribute to interactions with the viral RNA, polymerase, membranes and cellular proteins. The larger protein VPgs from potyviruses and noroviruses/caliciviruses may also show some areas of similar properties to these small peptides.
Collapse
Affiliation(s)
- Catherine H Schein
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
56
|
Luis-Ravelo D, Fumagallo-Reading F, Febles-Casquero A, Lopez-Fernandez J, Marcellino DJ, Gonzalez-Hernandez T. Dopamine Receptor D3 Induces Transient, mTORC1-Dependent Autophagy That Becomes Persistent, AMPK-Mediated, and Neuroprotective in Experimental Models of Huntington's Disease. Cells 2025; 14:652. [PMID: 40358175 PMCID: PMC12071662 DOI: 10.3390/cells14090652] [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: 03/18/2025] [Revised: 04/21/2025] [Accepted: 04/27/2025] [Indexed: 05/15/2025] Open
Abstract
Huntington disease's (HD) is a neurodegenerative disorder caused by the expansion of a polyglutamine region (PolyQ) within the huntingtin protein (HTT). Mutated huntingtin (mHTT) is cytotoxic, particularly for striatal medium spiny neurons (MSNs), whose degeneration is the hallmark of HD. Autophagy inducers currently available promote the clearance of toxic proteins. However, due to their low selectivity and the possibility that prolonged autophagy hampers essential processes in unaffected cells, researchers have questioned their benefits in neurodegenerative diseases. Since MSNs express dopamine receptors D2 (DRD2) and D3 (DRD3) and DRD2/DRD3 agonists may activate autophagy, here, we explored how healthy and mHTT-challenged cells respond to prolonged DRD2/DRD3 agonist treatment. Autophagy activation and its effects on mHTT/polyQ clearance were studied in R6/1 mice (a genetic model of HD), their wild-type littermates, and DRD2- and DRD3-HEK cells expressing a pathogenic (Q74) and a non-pathogenic (Q23) polyQ fragment of mHTT treated with the DRD2/DRD3 agonist pramipexole. Two forms of DRD3-mediated autophagy were found: a transient mTORC1-dependent in WT mice and Q23-DRD3-HEK cells and a persistent AMPK-ULK1-activated in R6/1 mice and Q74-DRD3-HEK cells. This also promoted a robust clearance of soluble mHTT/polyQ and neuroprotection in striatal neurons and DRD3-HEK cells. The findings indicate that DRD3-induced autophagy may be a safe, disease-modifying intervention in HD patients.
Collapse
Affiliation(s)
- Diego Luis-Ravelo
- Institute of Biomedical Technologies, University of La Laguna, 38200 Tenerife, Spain; (D.L.-R.); (F.F.-R.)
| | - Felipe Fumagallo-Reading
- Institute of Biomedical Technologies, University of La Laguna, 38200 Tenerife, Spain; (D.L.-R.); (F.F.-R.)
| | - Alejandro Febles-Casquero
- Department of Basic Medical Sciences, Faculty of Medicine, University of La Laguna, 38200 Tenerife, Spain
| | - Jonathan Lopez-Fernandez
- Institute of Biomedical Technologies, University of La Laguna, 38200 Tenerife, Spain; (D.L.-R.); (F.F.-R.)
- Department of Basic Medical Sciences, Faculty of Medicine, University of La Laguna, 38200 Tenerife, Spain
| | - Daniel J. Marcellino
- Department of Medical and Translational Biology, Umeå University, 901 87 Umeå, Sweden;
| | - Tomas Gonzalez-Hernandez
- Institute of Biomedical Technologies, University of La Laguna, 38200 Tenerife, Spain; (D.L.-R.); (F.F.-R.)
- Department of Basic Medical Sciences, Faculty of Medicine, University of La Laguna, 38200 Tenerife, Spain
| |
Collapse
|
57
|
Parizad R, Batta A, Hatwal J, Taban-Sadeghi M, Mohan B. Emerging risk factors for heart failure in younger populations: A growing public health concern. World J Cardiol 2025; 17:104717. [PMID: 40308622 PMCID: PMC12038706 DOI: 10.4330/wjc.v17.i4.104717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 03/07/2025] [Accepted: 04/01/2025] [Indexed: 04/21/2025] Open
Abstract
Heart failure (HF) is a growing public health concern, with an increasing incidence among younger populations. Traditionally, HF was considered a condition primarily affecting the elderly, but of late, emerging evidence hints at a rapidly rising HF incidence in youth in the past 2 decades. HF in youth has been linked to a complex interaction between emerging risk factors, such as metabolic syndrome, environmental exposures, genetic predispositions, and lifestyle behaviors. This review examines these evolving determinants, including substance abuse, autoimmune diseases, and the long-term cardiovascular effects of coronavirus disease 2019, which disproportionately affect younger individuals. Through a comprehensive analysis, the study highlights the importance of early detection, targeted prevention strategies, and multidisciplinary management approaches to address this alarming trend. Promoting awareness and integrating age-specific interventions could significantly reduce the burden of HF and improve long-term outcomes among younger populations.
Collapse
Affiliation(s)
- Razieh Parizad
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz 51656-87386, Iran
| | - Akash Batta
- Department of Cardiology, Dayanand Medical College and Hospital, Ludhiana 141001, Punjab, India.
| | - Juniali Hatwal
- Department of Internal Medicine, Post Graduate Institute of Medical Education & Research, Chandigarh 160012, India
| | | | - Bishav Mohan
- Department of Cardiology, Dayanand Medical College and Hospital, Ludhiana 141001, Punjab, India
| |
Collapse
|
58
|
Lufrano D, Gong C, Cecarini V, Cuccioloni M, Bonfili L, Sturaro C, Bettegazzi B, Ruzza C, Perelló M, Angeletti M, Eleuteri AM. An Insight into Neuronal Processing of Ghrelin: Effects of a Bioactive Ghrelin Derivative on Proteolytic Pathways and Mitophagy. Mol Neurobiol 2025:10.1007/s12035-025-04976-5. [PMID: 40285938 DOI: 10.1007/s12035-025-04976-5] [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: 12/31/2024] [Accepted: 04/16/2025] [Indexed: 04/29/2025]
Abstract
Protein homeostasis (proteostasis) is preserved by an orchestrated network of molecular mechanisms that regulate protein synthesis, folding, and degradation, ensuring cellular integrity and function. Proteostasis declines with age and is related to pathologies such as neurodegenerative diseases and cardiac disorders, which are accompanied by the accumulation of toxic protein aggregates. In this context, therapeutic strategies enhancing the two primary degradative systems involved in the cellular clearance of those abnormal proteins, namely ubiquitin-proteasome system and autophagy-lysosomal pathway, represent a promising approach to counteract the collapse of proteostasis in such pathological conditions. In this work, we explored the processing of ghrelin, a pleiotropic peptide hormone linked to energy metabolism and higher brain functions, which is reported to modulate the protein degradative mechanisms. According to our data, ghrelin is processed by serine hydrolases secreted into the conditioned medium of SH-SY5Y neuroblastoma cell line, commonly used in neurotoxicology and neuroscience research. Ghrelin processing leads to the formation of a shorter peptide (ghrelin(1-11)) that stimulates both the cell proteasome system and autophagy-lysosomal pathway, encompassing the selective autophagy of mitochondria. Our findings suggest that ghrelin processing may contribute to the maintenance of neuronal proteostasis.
Collapse
Affiliation(s)
- Daniela Lufrano
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032, Camerino, Italy.
- Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional La Plata (UNLP), CONICET, B1900 AVW, La Plata, Argentina.
| | - Chunmei Gong
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032, Camerino, Italy
- Key Laboratory of Tropical Translational Medicine of the Ministry of Education, Hainan Medical University, 571199, Haikou, China
| | - Valentina Cecarini
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032, Camerino, Italy
| | - Massimiliano Cuccioloni
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032, Camerino, Italy
| | - Laura Bonfili
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032, Camerino, Italy
| | - Chiara Sturaro
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121, Ferrara, Italy
| | | | - Chiara Ruzza
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121, Ferrara, Italy
| | - Mario Perelló
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional La Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Comisión de Investigaciones Científicas de La Provincia de Buenos Aires (CIC-PBA), B1906 APM, La Plata, Argentina
| | - Mauro Angeletti
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032, Camerino, Italy
| | - Anna Maria Eleuteri
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032, Camerino, Italy.
| |
Collapse
|
59
|
Seo MK, Kim H, Choi AJ, Seog DH, Kho WG, Park SW, Lee JG. Effects of tianeptine on mTORC1-mediated neuronal autophagy in primary rat hippocampal neurons under nutrient deprivation. Sci Rep 2025; 15:14488. [PMID: 40280952 PMCID: PMC12032415 DOI: 10.1038/s41598-025-92988-5] [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: 12/07/2024] [Accepted: 03/04/2025] [Indexed: 04/29/2025] Open
Abstract
The aim of this study was to investigate the effects of the antidepressant tianeptine on the mechanistic target of rapamycin complex 1(mTORC1)-mediated autophagy pathway in primary hippocampal neurons exposed to B27-deprived conditions. When primary hippocampal neurons were treated with tianeptine at doses of 1, 10, 50, and 100 µM for 3 days under B27-deprived conditions, we observed that it activated autophagy and increased the formation of autophagosomes through the upregulation of autophagic proteins, including autophagy-activating kinase 1 (ULK1), Beclin 1, LC3B-II/I, and p62. And at a concentration of 100 µM tianeptine, the decrease in mTORC1 phosphorylation induced by B27 deprivation was significantly reversed. Changes in the expression of autophagic proteins induced by B27 deprivation were reversed by tianeptine treatment in a concentration-dependent manner, and tianeptine significantly reduced the increase in LC3B membrane number induced by B27 deprivation, an effect that was blocked by pretreatment with rapamycin. In conclusion, tianeptine attenuated the activity of mTORC1-mediated autophagy in primary rat hippocampal neurons under B27-deprived conditions. These results may suggest a novel mechanism by which tianeptine may affect autophagy in neurons.
Collapse
Affiliation(s)
- Mi Kyoung Seo
- Paik Institute for Clinical Research, Inje University, Busan, 47392, Republic of Korea
- Department of Convergence Biomedical Science, College of Medicine, Inje University, Busan, 47392, Republic of Korea
| | - Hyewon Kim
- Department of Psychiatry, College of Medicine, Haeundae Paik Hospital, Inje University, Busan, 48108, Republic of Korea
| | - Ah Jeong Choi
- Paik Institute for Clinical Research, Inje University, Busan, 47392, Republic of Korea
| | - Dae-Hyun Seog
- Department of Convergence Biomedical Science, College of Medicine, Inje University, Busan, 47392, Republic of Korea
- Department of Biochemistry, College of Medicine, Inje University, Busan, 47392, Republic of Korea
- Dementia and Neurodegenerative Disease Research Center, College of Medicine, Inje University, Busan, 47392, Republic of Korea
| | - Weon-Gyu Kho
- Paik Institute for Clinical Research, Inje University, Busan, 47392, Republic of Korea
- Department of Parasitology, College of Medicine, Inje University, Busan, 47392, Republic of Korea
| | - Sung Woo Park
- Paik Institute for Clinical Research, Inje University, Busan, 47392, Republic of Korea.
- Department of Convergence Biomedical Science, College of Medicine, Inje University, Busan, 47392, Republic of Korea.
| | - Jung Goo Lee
- Paik Institute for Clinical Research, Inje University, Busan, 47392, Republic of Korea.
- Department of Psychiatry, College of Medicine, Haeundae Paik Hospital, Inje University, Busan, 48108, Republic of Korea.
| |
Collapse
|
60
|
Tetteh P, Danso EK, Osei-Wusu S, Yeboah-Manu D, Asare P. The role of metformin in tuberculosis control among TB and diabetes mellitus comorbid individuals. Front Microbiol 2025; 16:1549572. [PMID: 40351311 PMCID: PMC12062004 DOI: 10.3389/fmicb.2025.1549572] [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: 12/21/2024] [Accepted: 04/07/2025] [Indexed: 05/14/2025] Open
Abstract
Tuberculosis (TB) is a major global health concern, and its control is particularly hindered in patients with comorbidities such as type 2 diabetes mellitus (TBDM). Metformin, a commonly prescribed antidiabetic medication, has gained attention because of its potential role in improving TB treatment outcomes in patients with TBDM. This review aims to assess the role of metformin in TB case management among TBDM comorbid individuals. By reviewing the available literature, we aimed to explore the potential benefits, complications, mechanisms, and considerations surrounding metformin use as an adjunctive therapy for TB treatment. The findings of this review will shed light on current understanding and highlight areas for further investigation.
Collapse
Affiliation(s)
| | | | | | | | - Prince Asare
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| |
Collapse
|
61
|
Zou Y, Zou Y, Yang B, Qiao J, Liu S, Zhao C, Shi K, Yu Y, Li S, Chen S. Effects of bacterial fertilizer and soil amendment on Spuriopinella brachycarpa (Kom.) Kitag. growth and soil microbiota. Front Microbiol 2025; 16:1560982. [PMID: 40351309 PMCID: PMC12061978 DOI: 10.3389/fmicb.2025.1560982] [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: 01/15/2025] [Accepted: 03/27/2025] [Indexed: 05/14/2025] Open
Abstract
Introduction Spuriopinella brachycarpa (Kom.) Kitag. is a nutritious wild vegetable, but its quality deteriorates during artificial cultivation due to soil condition alterations. Microbial fertilizers and soil amendments hold promise for improving cultivation outcomes, yet their combined effects remain under - explored. Methods A field experiment was conducted with seven treatments, including a control (CK) and six combinations of Trichoderma harzianum, Bacillus subtilis, and earthworm polysaccharide. Plant samples were analyzed for yield, quality, and root architecture, while soil samples were tested for fertility and microbial community characteristics. Results Treatments T3 (dual bacterial fertilizers) and T6 (dual bacterial fertilizers + soil amendment) significantly enhanced yield, nutritional quality, and root development. T6 also maintained high soil fertility and optimized the soil microbial community in terms of richness, diversity, and beneficial species abundance. Discussion The positive effects of T3 and T6 are likely due to the synergy between the bacterial fertilizers and the soil amendment, which improves nutrient cycling, soil structure, and microbial functions. However, the study has limitations, such as the need for long - term research and more in - depth exploration of microbial functions.
Collapse
Affiliation(s)
- Yue Zou
- College of Horticulture, Jilin Agricultural University, Changchun, China
| | - Yan Zou
- College of Horticulture, Jilin Agricultural University, Changchun, China
| | - Baiming Yang
- College of Horticulture, Jilin Agricultural University, Changchun, China
| | - Jianlei Qiao
- College of Horticulture, Jilin Agricultural University, Changchun, China
| | - Shuang Liu
- College of Horticulture, Jilin Agricultural University, Changchun, China
| | - Chunbo Zhao
- College of Horticulture, Jilin Agricultural University, Changchun, China
| | - Kun Shi
- Liaoyuan Sino-German Modern Agriculture Co., Ltd., Dongliao, China
| | - Yue Yu
- College of Horticulture, Jilin Agricultural University, Changchun, China
| | - Shuang Li
- Teaching and Research Base Management Office, Jilin Agricultural University, Changchun, China
| | - Shanshan Chen
- College of Horticulture, Jilin Agricultural University, Changchun, China
| |
Collapse
|
62
|
Gerardo H, Lourenço T, Torres J, Ferreira M, Aveleira C, Simões S, Ferreira L, Cavadas C, Oliveira PJ, Teixeira J, Grãos M. Extracellular matrix mechanical cues (dys)regulate metabolic redox homeostasis due to impaired autophagic flux. Eur J Clin Invest 2025:e70051. [PMID: 40280877 DOI: 10.1111/eci.70051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 04/07/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Extracellular matrix (ECM) stiffness is increasingly recognized as a critical regulator of cellular behaviour, governing processes such as proliferation, differentiation, and metabolism. Neurodegenerative diseases are characterized by mitochondrial dysfunction, oxidative stress, impaired autophagy, and progressive softening of the brain tissue, yet research into how mechanical cues influence cellular metabolism in this context remains scarce. MATERIALS AND METHODS In this study, we evaluated the long-term effects of brain-compliant, soft ECM on mitochondrial bioenergetics, redox balance, and autophagic capacity in human neuroblastoma (SH-SY5Y) and mouse hippocampal (HT22) cell lines, as well as primary mouse neurons. RESULTS We observed that prolonged exposure to soft ECM does not impact cell proliferative capacity of neuronal cells but results in mitochondrial bioenergetic dysfunction, redox imbalance, and disrupted autophagic flux. These findings were consistently validated across both human and mouse neuronal cells. Our data indicate a decreased maximal autophagic capacity in cells exposed to long-term soft ECM, potentially due to an imbalance in autophagosome formation and degradation, as demonstrated by decreased LC3 II levels following chloroquine-induced autophagic flux inhibition. This impairment in autophagy was coupled with increased cellular oxidative stress, further indicating metabolic alterations. CONCLUSIONS These findings emphasize the critical role of ECM stiffness in regulating neuronal cell metabolism and suggest that prolonged exposure to soft ECM may mimic key aspects of neurodegenerative disease pathology, thereby enhancing the physiological relevance of in vitro models. This study underscores the necessity for further research into ECM mechanics as a contributing factor in neurodegenerative disease progression and as a potential target for therapeutic strategies.
Collapse
Affiliation(s)
- Heloísa Gerardo
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Tânia Lourenço
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Júlio Torres
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research PDBEB - Doctoral Programme in Experimental Biology and Biomedicine, University of Coimbra, Coimbra, Portugal
| | - Manuela Ferreira
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Célia Aveleira
- CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Multidisciplinary Institute of Aging - MIA, Coimbra, Portugal
| | - Susana Simões
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Lino Ferreira
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Cláudia Cavadas
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Paulo J Oliveira
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - José Teixeira
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Department of Life Science (DCV), Faculty of Science and Technology, University of Coimbra, Coimbra, Portugal
| | - Mário Grãos
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
63
|
Zhou N, Chen J, Hu M, Wen N, Cai W, Li P, Zhao L, Meng Y, Zhao D, Yang X, Liu S, Huang F, Zhao C, Feng X, Jiang Z, Xie E, Pan H, Cen Z, Chen X, Luo W, Tang B, Min J, Wang F, Yang J, Xu H. SLC7A11 is an unconventional H + transporter in lysosomes. Cell 2025:S0092-8674(25)00406-4. [PMID: 40280132 DOI: 10.1016/j.cell.2025.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 01/22/2025] [Accepted: 04/02/2025] [Indexed: 04/29/2025]
Abstract
Lysosomes maintain an acidic pH of 4.5-5.0, optimal for macromolecular degradation. Whereas proton influx is produced by a V-type H+ ATPase, proton efflux is mediated by a fast H+ leak through TMEM175 channels, as well as an unidentified slow pathway. A candidate screen on an orphan lysosome membrane protein (OLMP) library enabled us to discover that SLC7A11, the protein target of the ferroptosis-inducing compound erastin, mediates a slow lysosomal H+ leak through downward flux of cystine and glutamate, two H+ equivalents with uniquely large but opposite concentration gradients across lysosomal membranes. SLC7A11 deficiency or inhibition caused lysosomal over-acidification, reduced degradation, accumulation of storage materials, and ferroptosis, as well as facilitated α-synuclein aggregation in neurons. Correction of abnormal lysosomal acidity restored lysosome homeostasis and prevented ferroptosis. These studies have revealed an unconventional H+ transport conduit that is integral to lysosomal flux of protonatable metabolites to regulate lysosome function, ferroptosis, and Parkinson's disease (PD) pathology.
Collapse
Affiliation(s)
- Nan Zhou
- New Cornerstone Science Laboratory and Liangzhu Laboratory, the Second Affiliated Hospital and School of Basic Medical Sciences, Zhejiang University, Hangzhou, China; Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Jingzhi Chen
- New Cornerstone Science Laboratory and Liangzhu Laboratory, the Second Affiliated Hospital and School of Basic Medical Sciences, Zhejiang University, Hangzhou, China; Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Meiqin Hu
- New Cornerstone Science Laboratory and Liangzhu Laboratory, the Second Affiliated Hospital and School of Basic Medical Sciences, Zhejiang University, Hangzhou, China.
| | - Na Wen
- New Cornerstone Science Laboratory and Liangzhu Laboratory, the Second Affiliated Hospital and School of Basic Medical Sciences, Zhejiang University, Hangzhou, China; Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Weijie Cai
- New Cornerstone Science Laboratory and Liangzhu Laboratory, the Second Affiliated Hospital and School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
| | - Ping Li
- New Cornerstone Science Laboratory and Liangzhu Laboratory, the Second Affiliated Hospital and School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
| | - Liding Zhao
- New Cornerstone Science Laboratory and Liangzhu Laboratory, the Second Affiliated Hospital and School of Basic Medical Sciences, Zhejiang University, Hangzhou, China; Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yaping Meng
- New Cornerstone Science Laboratory and Liangzhu Laboratory, the Second Affiliated Hospital and School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
| | - Dongdong Zhao
- New Cornerstone Science Laboratory and Liangzhu Laboratory, the Second Affiliated Hospital and School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
| | - Xiaotong Yang
- New Cornerstone Science Laboratory and Liangzhu Laboratory, the Second Affiliated Hospital and School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
| | - Siyu Liu
- New Cornerstone Science Laboratory and Liangzhu Laboratory, the Second Affiliated Hospital and School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
| | - Fangqian Huang
- New Cornerstone Science Laboratory and Liangzhu Laboratory, the Second Affiliated Hospital and School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
| | - Cheng Zhao
- New Cornerstone Science Laboratory and Liangzhu Laboratory, the Second Affiliated Hospital and School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
| | - Xinghua Feng
- New Cornerstone Science Laboratory and Liangzhu Laboratory, the Second Affiliated Hospital and School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
| | - Zikai Jiang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Enjun Xie
- The Second Affiliated Hospital & the First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China
| | - Hongxu Pan
- Department of Neurology & National Clinical Research Centre for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhidong Cen
- Department of Neurology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinhui Chen
- Department of Neurology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Luo
- Department of Neurology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Beisha Tang
- Department of Neurology & National Clinical Research Centre for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Junxia Min
- The Second Affiliated Hospital & the First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China
| | - Fudi Wang
- The Second Affiliated Hospital & the First Affiliated Hospital, Institute of Translational Medicine, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China
| | - Junsheng Yang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Haoxing Xu
- New Cornerstone Science Laboratory and Liangzhu Laboratory, the Second Affiliated Hospital and School of Basic Medical Sciences, Zhejiang University, Hangzhou, China; Institute of Fundamental and Transdisciplinary Research and The State Key Lab of Brain-Machine Intelligence, Zhejiang University, Hangzhou, China; Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
64
|
NAKASHIMA MOEKA, SUGA NAOKO, FUKUMOTO AKARI, YOSHIKAWA SAYURI, MATSUDA SATORU. Promising roles of vitamin D receptor and APRO family proteins for the development of cancer stem cells targeted malignant tumor therapy. Oncol Res 2025; 33:1007-1017. [PMID: 40296902 PMCID: PMC12034005 DOI: 10.32604/or.2025.059657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 01/13/2025] [Indexed: 04/30/2025] Open
Abstract
Malignant tumors are heterogeneous diseases characterized by uncontrolled cell proliferation, invasion, metastasis, and/or recurrence of their malignancies. In particular, cancer stem cells (CSCs) within these tumors might be responsible for the property of invasiveness and/or therapies-resistance. CSCs are a self-renewing, awfully tumorigenic subpopulation of cancer cells, which are notorious for strong chemoresistance and are frequently responsible the aggravated invasion, metastasis, and/or recurrence. Developing targeting therapies against CSCs, therefore, may be deliberated a more encouraging mission for the greater cancer therapy. Innovation for a more potent anti-CSC treatment has been required as soon as possible. Interestingly, vitamin D could modulate the inflammatory condition of the tumor microenvironment (TME) by successfully affecting CSCs, which has an imperative role in determining the malignant phenotype of CSCs. In addition, vitamin D may also contribute to the regulation of the malignant behaviors of CSCs. Consistently, vitamin D could have potential applications for the significant inhibition of several tumor growths within various cancer therapies. The biological significance of vitamin D for CSCs regulation may be involved in the function of APRO family proteins. Therefore, vitamin D could be one of the innovative therapeutic modalities for the development of novel CSCs related tumor therapies.
Collapse
Affiliation(s)
- MOEKA NAKASHIMA
- Department of Food Science and Nutrition, Nara Women’s University, Kita-Uoya Nishimachi, Nara, 630-8506, Japan
| | - NAOKO SUGA
- Department of Food Science and Nutrition, Nara Women’s University, Kita-Uoya Nishimachi, Nara, 630-8506, Japan
| | - AKARI FUKUMOTO
- Department of Food Science and Nutrition, Nara Women’s University, Kita-Uoya Nishimachi, Nara, 630-8506, Japan
| | - SAYURI YOSHIKAWA
- Department of Food Science and Nutrition, Nara Women’s University, Kita-Uoya Nishimachi, Nara, 630-8506, Japan
| | - SATORU MATSUDA
- Department of Food Science and Nutrition, Nara Women’s University, Kita-Uoya Nishimachi, Nara, 630-8506, Japan
| |
Collapse
|
65
|
Pan C, Cheng C, Zhong S, Li S, Tan W, Yao Y. In vitro study on the promotion of osteogenic differentiation by mitochondrial-derived vesicles through activation of inflammation and reprogramming of metabolic pathways. J Orthop Surg Res 2025; 20:388. [PMID: 40247396 PMCID: PMC12007352 DOI: 10.1186/s13018-025-05749-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 03/22/2025] [Indexed: 04/19/2025] Open
Abstract
The secretion of mitochondrial-derived vesicles (MDVs) has been found to increase during osteogenic differentiation, but their role in intercellular communication and osteogenic promotion remains unclear. In this study, we extracted translocase of outer mitochondrial membrane 20 (Tomm20) + MDVs from bone marrow stromal cells (BMSCs) at different osteogenic culture days using differential centrifugation and immunoprecipitation, then co-cultured them with BMSCs to assess osteogenic differentiation, immune response and metabolic levels. The results showed that osteogenic differentiation enhances MDVs' secretion and their mitochondrial DNA (mtDNA) content. In promoting osteogenic differentiation ability, osteogenic-induced MDVs (MDV-OMs, especially MDV-OM14 and MDV-OM21) significantly enhance mineralization with OD values 1.37-fold and 1.32-fold higher than those of MDV-OM7 (p < 0.05) after 21 days, respectively. However, these MDVs containing mtDNA activate immune responses by upregulating cGas, Sting, Caspase-9, Il-6, and Tnf-a mRNA levels, inducing cell apoptosis and oxidative stress. In addition, MDVs containing mitochondrial components also have metabolic regulatory functions. Metabolic level detection revealed that MDV-OMs downregulate lactate, promote tricarboxylic acid cycle (TCA) enzyme expression, and increase mitochondrial membrane potential. Among these MDVs, MDV-OM7, induced for 7 days, shows osteogenic function without strong immune response, possibly related to metabolic reprogramming. This study highlights the potential of osteogenic-induced MDVs for bone regeneration, cGAS-STING activation, and metabolic enhancement, and are expected to be used for the treatment of diseases such as tissue damage.
Collapse
Affiliation(s)
- Chun Pan
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
- Institute of Genomic Medicine, College of Pharmacy, Jinan University, Guangzhou, 511436, China
| | - Cheng Cheng
- Institute of Genomic Medicine, College of Pharmacy, Jinan University, Guangzhou, 511436, China
- Department of Paediatric Orthopaedics, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, 510632, China
| | - Shu Zhong
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Shiyu Li
- Department of Microbiology and Immunology, College of Basic Medicine and Public Hygiene, Jinan University, Guangzhou, 510632, China.
| | - Wei Tan
- Department of Paediatric Orthopaedics, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, 510632, China.
| | - Yachao Yao
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China.
| |
Collapse
|
66
|
Al-Bari MAA, Davamani F, Bhatnagar P, Eid N. Plantamajoside mitigates endoplasmic reticulum stress-mediated pancreatic β-cell apoptosis in type 2 diabetes via DNAJC1 upregulation. World J Diabetes 2025; 16:104241. [PMID: 40236854 PMCID: PMC11947917 DOI: 10.4239/wjd.v16.i4.104241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/14/2025] [Accepted: 01/22/2025] [Indexed: 02/28/2025] Open
Abstract
Diabetes mellitus (DM) is a metabolic disorder characterized by persistent hyperglycemia and other symptoms, which pose significant challenges to individual health, life expectancy, and public healthcare systems. The escalating global prevalence of diabetes underscores the need for innovative therapeutic interventions. In this article, we critically comment on the study by Wang et al, published in the World Journal of Diabetes, which elucidates the therapeutic potential of Plantamajoside (PMS) in type 2 DM (T2DM) management. The authors provide evidence for the mechanism of action of PMS in T2DM models, demonstrating prevention of endoplasmic reticulum stress and apoptosis of pancreatic β-cells via activation of DNAJC1. This manuscript provides a brief review of the pathogenesis of T2DM, explores the various roles of PMS in disease therapy in addition to the DNAJC-related apoptotic and autophagic functions, critically evaluates the experimental approaches employed by Wang et al, and provides recommendations for advancing future research.
Collapse
Affiliation(s)
| | - Fabian Davamani
- Division of Applied Biomedical Sciences and Biotechnology, School of Health Sciences, IMU University, Kuala Lumpur 57000, Malaysia
| | - Payal Bhatnagar
- Department of Pharmaceutical Technology, School of Pharmacy, IMU University, Kuala Lumpur 57000, Malaysia
| | - Nabil Eid
- Department of Anatomy, Division of Human Biology, School of Medicine, IMU University, Kuala Lumpur 57000, Malaysia
| |
Collapse
|
67
|
Faris ME, Khaled MB, Chtourou H, Kalam F, Abdelrahim DN, BaHammam AS. Editorial: The model of Ramadan diurnal intermittent fasting: unraveling the health implications, volume III. Front Nutr 2025; 12:1586573. [PMID: 40290664 PMCID: PMC12023006 DOI: 10.3389/fnut.2025.1586573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Accepted: 03/21/2025] [Indexed: 04/30/2025] Open
Affiliation(s)
- MoezAlIslam Ezzat Faris
- Department of Clinical Nutrition and Dietetics, Applied Science Private University, Amman, Jordan
| | - Meghit Boumediene Khaled
- Faculté des Sciences de la Nature et de la Vie, Univeristé Djillali Liabes Sidi Bel Abbès, Sidi Bel Abbes, Algeria
| | - Hamdi Chtourou
- High Institute of Sport and Physical Education of Sfax, Sfax, Tunisia
| | - Faiza Kalam
- Division of Cancer Prevention and Control, Department of Internal Medicine, The Ohio State University, Columbus, OH, United States
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | | | - Ahmed S. BaHammam
- University Sleep Disorders Center, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
68
|
Hou S, Wang C, Ma X, Zhao J, Wang J, Fang Y, Liu H, Ding H, Guo J, Lu W. Methylmercury Chloride Exposure Affects Oocyte Maturation Through AMPK/mTOR-Mediated Mitochondrial Autophagy. Int J Mol Sci 2025; 26:3603. [PMID: 40332119 PMCID: PMC12026530 DOI: 10.3390/ijms26083603] [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: 03/02/2025] [Revised: 03/28/2025] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
Mercury, a prevalent heavy metal, negatively impacts oocyte maturation. However, the exact mechanism by which methylmercury chloride (MMC) affects this process remains elusive. The present study found that MMC administration triggered meiotic failure in oocytes by disrupting cumulus cell expansion, leading to compromised spindle apparatus and altered chromosomal architecture, which are crucial for oocyte development. This disruption is characterized by abnormal microtubule organization and defective chromosome alignment. Additionally, MMC exposure caused oxidative stress-induced apoptosis due to mitochondrial dysfunction, as indicated by decreased mitochondrial membrane potential, mitochondrial content, mitochondrial DNA copy number, and adenosine triphosphate levels. Proteomic analysis identified 97 differentially expressed proteins, including P62, an autophagy marker. Our results confirmed that MMC induced autophagy, particularly through the hyperactivation of the mitochondrial autophagy to remove damaged and normal mitochondria. The mitochondrial reactive oxygen species (ROS) scavenger Mito-TEMPO alleviated oxidative stress and mitochondrial autophagy levels, suggesting that mitochondrial ROS initiates this autophagic response. Notably, MMC activates mitochondrial autophagy via the monophosphate-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR) signal pathway due to mitochondrial dysfunction. In vivo studies in mice revealed that MMC exposure decreased reproductive performance, attributed to excessive mitochondrial autophagy leading to reduced oocyte quality. Overall, these findings demonstrate that MMC exposure impairs oocyte maturation via the hyperactivation of mitochondrial autophagy induced by mitochondrial dysfunction.
Collapse
Affiliation(s)
- Shengkui Hou
- Key Laboratory of the Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Changchun 130117, China; (S.H.); (X.M.); (J.Z.); (J.W.); (Y.F.); (H.L.); (H.D.)
- Jilin Provincial International Joint Research Center of Animal Breeding and Reproduction Technology, Jilin Agricultural University, Changchun 130117, China
- Jilin Provincial Key Laboratory of Beef Cattle Germplasm Resources Conservation and Utilization, Jilin Agricultural University, Changchun 130117, China
| | - Caiyu Wang
- Key Laboratory of the Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Changchun 130117, China; (S.H.); (X.M.); (J.Z.); (J.W.); (Y.F.); (H.L.); (H.D.)
- Jilin Provincial International Joint Research Center of Animal Breeding and Reproduction Technology, Jilin Agricultural University, Changchun 130117, China
- Jilin Provincial Key Laboratory of Beef Cattle Germplasm Resources Conservation and Utilization, Jilin Agricultural University, Changchun 130117, China
| | - Xin Ma
- Key Laboratory of the Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Changchun 130117, China; (S.H.); (X.M.); (J.Z.); (J.W.); (Y.F.); (H.L.); (H.D.)
- Jilin Provincial International Joint Research Center of Animal Breeding and Reproduction Technology, Jilin Agricultural University, Changchun 130117, China
- Jilin Provincial Key Laboratory of Beef Cattle Germplasm Resources Conservation and Utilization, Jilin Agricultural University, Changchun 130117, China
| | - Jing Zhao
- Key Laboratory of the Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Changchun 130117, China; (S.H.); (X.M.); (J.Z.); (J.W.); (Y.F.); (H.L.); (H.D.)
- Jilin Provincial International Joint Research Center of Animal Breeding and Reproduction Technology, Jilin Agricultural University, Changchun 130117, China
- Jilin Provincial Key Laboratory of Beef Cattle Germplasm Resources Conservation and Utilization, Jilin Agricultural University, Changchun 130117, China
| | - Jun Wang
- Key Laboratory of the Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Changchun 130117, China; (S.H.); (X.M.); (J.Z.); (J.W.); (Y.F.); (H.L.); (H.D.)
- Jilin Provincial International Joint Research Center of Animal Breeding and Reproduction Technology, Jilin Agricultural University, Changchun 130117, China
- Jilin Provincial Key Laboratory of Beef Cattle Germplasm Resources Conservation and Utilization, Jilin Agricultural University, Changchun 130117, China
| | - Yi Fang
- Key Laboratory of the Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Changchun 130117, China; (S.H.); (X.M.); (J.Z.); (J.W.); (Y.F.); (H.L.); (H.D.)
- Jilin Provincial International Joint Research Center of Animal Breeding and Reproduction Technology, Jilin Agricultural University, Changchun 130117, China
- Jilin Provincial Key Laboratory of Beef Cattle Germplasm Resources Conservation and Utilization, Jilin Agricultural University, Changchun 130117, China
| | - Hongyu Liu
- Key Laboratory of the Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Changchun 130117, China; (S.H.); (X.M.); (J.Z.); (J.W.); (Y.F.); (H.L.); (H.D.)
- Jilin Provincial International Joint Research Center of Animal Breeding and Reproduction Technology, Jilin Agricultural University, Changchun 130117, China
- Jilin Provincial Key Laboratory of Beef Cattle Germplasm Resources Conservation and Utilization, Jilin Agricultural University, Changchun 130117, China
| | - He Ding
- Key Laboratory of the Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Changchun 130117, China; (S.H.); (X.M.); (J.Z.); (J.W.); (Y.F.); (H.L.); (H.D.)
- Jilin Provincial International Joint Research Center of Animal Breeding and Reproduction Technology, Jilin Agricultural University, Changchun 130117, China
- Jilin Provincial Key Laboratory of Beef Cattle Germplasm Resources Conservation and Utilization, Jilin Agricultural University, Changchun 130117, China
| | - Jing Guo
- Key Laboratory of the Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Changchun 130117, China; (S.H.); (X.M.); (J.Z.); (J.W.); (Y.F.); (H.L.); (H.D.)
- Jilin Provincial International Joint Research Center of Animal Breeding and Reproduction Technology, Jilin Agricultural University, Changchun 130117, China
- Jilin Provincial Key Laboratory of Beef Cattle Germplasm Resources Conservation and Utilization, Jilin Agricultural University, Changchun 130117, China
| | - Wenfa Lu
- Key Laboratory of the Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Changchun 130117, China; (S.H.); (X.M.); (J.Z.); (J.W.); (Y.F.); (H.L.); (H.D.)
- Jilin Provincial International Joint Research Center of Animal Breeding and Reproduction Technology, Jilin Agricultural University, Changchun 130117, China
- Jilin Provincial Key Laboratory of Beef Cattle Germplasm Resources Conservation and Utilization, Jilin Agricultural University, Changchun 130117, China
| |
Collapse
|
69
|
Zhao K, Chan ITC, Tse EHY, Xie Z, Cheung TH, Zeng YA. Autophagy in adult stem cell homeostasis, aging, and disease therapy. CELL REGENERATION (LONDON, ENGLAND) 2025; 14:14. [PMID: 40208372 PMCID: PMC11985830 DOI: 10.1186/s13619-025-00224-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/16/2025] [Accepted: 02/23/2025] [Indexed: 04/11/2025]
Abstract
Autophagy is a crucial cellular process that facilitates the degradation of damaged organelles and protein aggregates, and the recycling of cellular components for the energy production and macromolecule synthesis. It plays an indispensable role in maintaining cellular homeostasis. Over recent decades, research has increasingly focused on the role of autophagy in regulating adult stem cells (SCs). Studies suggest that autophagy modulates various cellular processes and states of adult SCs, including quiescence, proliferation, self-renewal, and differentiation. The primary role of autophagy in these contexts is to sustain homeostasis, withstand stressors, and supply energy. Notably, the dysfunction of adult SCs during aging is correlated with a decline in autophagic activity, suggesting that autophagy is also involved in SC- and aging-associated disorders. Given the diverse cellular processes mediated by autophagy and the intricate mechanisms governing adult SCs, further research is essential to elucidate both universal and cell type-specific regulatory pathways of autophagy. This review discusses the role of autophagy in regulating adult SCs during quiescence, proliferation, self-renewal, and differentiation. Additionally, it summarizes the relationship between SC aging and autophagy, providing therapeutical insights into treating and ameliorating aging-associated diseases and cancers, and ultimately promoting longevity.
Collapse
Affiliation(s)
- Ke Zhao
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
- New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Indigo T C Chan
- Division of Life Science, Center for Stem Cell Research, State Key Laboratory of Molecular Neuroscience, Daniel and Mayce Yu Molecular Neuroscience Center, HKUST-Nan Fung Life Sciences Joint Laboratory, the Hong Kong University of Science and Technology, Hong Kong, China
| | - Erin H Y Tse
- Division of Life Science, Center for Stem Cell Research, State Key Laboratory of Molecular Neuroscience, Daniel and Mayce Yu Molecular Neuroscience Center, HKUST-Nan Fung Life Sciences Joint Laboratory, the Hong Kong University of Science and Technology, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Zhiyao Xie
- New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Tom H Cheung
- Division of Life Science, Center for Stem Cell Research, State Key Laboratory of Molecular Neuroscience, Daniel and Mayce Yu Molecular Neuroscience Center, HKUST-Nan Fung Life Sciences Joint Laboratory, the Hong Kong University of Science and Technology, Hong Kong, China.
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China.
| | - Yi Arial Zeng
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
- New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
70
|
Acosta Ingram D, Turkes E, Kim TY, Vo S, Sweeney N, Bonte MA, Rutherford R, Julian DL, Pan M, Marsh J, Argouarch AR, Wu M, Scharre DW, Bell EH, Honig LS, Vonsattel JP, Serrano GE, Beach TG, Karch CM, Kao AW, Hester ME, Han X, Fu H. GRAMD1B is a regulator of lipid homeostasis, autophagic flux and phosphorylated tau. Nat Commun 2025; 16:3312. [PMID: 40204713 PMCID: PMC11982250 DOI: 10.1038/s41467-025-58585-w] [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: 02/28/2025] [Accepted: 03/26/2025] [Indexed: 04/11/2025] Open
Abstract
Lipid dyshomeostasis and tau pathology are present in frontotemporal lobar degeneration (FTLD) and Alzheimer's disease (AD). However, the relationship between lipid dyshomeostasis and tau pathology remains unclear. We report that GRAM Domain Containing 1B (GRAMD1B), a nonvesicular cholesterol transporter, is increased in excitatory neurons of human neural organoids (HNOs) with the MAPT R406W mutation. Human FTLD, AD cases, and PS19 tau mice also have increased GRAMD1B expression. We show that overexpression of GRAMD1B increases levels of free cholesterol, lipid droplets, and impairs autophagy flux. Modulating GRAMD1B in iPSC-derived neurons also alters key autophagy-related components such as PI3K, phospho-AKT, and p62, as well as phosphorylated tau, and CDK5R1. Blocking GRAMD1B function decreases free cholesterol and lipid droplets. Knocking down GRAMD1B additionally reduces phosphorylated tau, and CDK5R1 expression. Our findings elucidate the role of GRAMD1B in the nervous system and highlight its relevance to FTLD and AD.
Collapse
Affiliation(s)
- Diana Acosta Ingram
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Emir Turkes
- UK Dementia Research Institute, UCL Queen Square Institute of Neurology, London, UK
| | - Tae Yeon Kim
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH, USA
- Biomedical Sciences Graduate Program, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Sheeny Vo
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Nicholas Sweeney
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Marie-Amandine Bonte
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Ryan Rutherford
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Dominic L Julian
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Meixia Pan
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Jacob Marsh
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrea R Argouarch
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Min Wu
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Douglas W Scharre
- Department of Neurology, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Erica H Bell
- Department of Neurology, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Lawrence S Honig
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Jean Paul Vonsattel
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | | | | | - Celeste M Karch
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Aimee W Kao
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Mark E Hester
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Xianlin Han
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Hongjun Fu
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH, USA.
- Chronic Brain Injury Program, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
71
|
Bradic I, Rewitz K. Steroid Signaling in Autophagy. J Mol Biol 2025:169134. [PMID: 40210154 DOI: 10.1016/j.jmb.2025.169134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/19/2025] [Accepted: 04/04/2025] [Indexed: 04/12/2025]
Abstract
Autophagy is a conserved cellular process essential for homeostasis and development that plays a central role in the degradation and recycling of cellular components. Recent studies reveal bidirectional interactions between autophagy and steroid-hormone signaling. Steroids are signaling molecules synthesized from cholesterol that regulate key physiological and developmental processes - including autophagic activity. Conversely, other work demonstrates that autophagy regulates steroid production by controlling the availability of precursor sterol substrate. Insights from Drosophila and mammalian models provide compelling evidence for the conservation of these mechanisms across species. In this review we explore how steroid hormones modulate autophagy in diverse tissues and contexts, such as metabolism and disease, and discuss advances in our understanding of autophagy's regulatory role in steroid hormone production. We examine the implications of these interactions for health and disease and offer perspectives on the potential for harnessing this functionality for addressing cholesterol-related disorders.
Collapse
Affiliation(s)
- Ivan Bradic
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense, Denmark
| | - Kim Rewitz
- Department of Biology, University of Copenhagen, 2100 Copenhagen O, Denmark.
| |
Collapse
|
72
|
Cai D, Li J, Peng Z, Fu R, Chen C, Liu F, Li Y, Su Y, Li C, Chen W. Interplay of Ferroptosis, Cuproptosis, Autophagy and Pyroptosis in Male Infertility: Molecular Crossroads and Therapeutic Opportunities. Int J Mol Sci 2025; 26:3496. [PMID: 40331931 PMCID: PMC12026609 DOI: 10.3390/ijms26083496] [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: 02/08/2025] [Revised: 04/01/2025] [Accepted: 04/04/2025] [Indexed: 05/08/2025] Open
Abstract
Male infertility is intricately linked to dysregulated cell death pathways, including ferroptosis, cuproptosis, pyroptosis, and autophagy. Ferroptosis, driven by iron-dependent lipid peroxidation through the Fenton reaction and inactivation of the GPX4/Nrf2/SLC7A11 axis, disrupts spermatogenesis under conditions of oxidative stress, environmental toxin exposure, or metabolic disorders. Similarly, cuproptosis-characterized by mitochondrial dysfunction and disulfide stress due to copper overload-exacerbates germ cell apoptosis via FDX1 activation and NADPH depletion. Pyroptosis, mediated by the NLRP3 inflammasome and gasdermin D, amplifies testicular inflammation and germ cell loss via IL-1β/IL-18 release, particularly in response to environmental insults. Autophagy maintains testicular homeostasis by clearing damaged organelles and proteins; however, its dysregulation impairs sperm maturation and compromises blood-testis barrier integrity. These pathways intersect through shared regulators; reactive oxygen species and mTOR modulate the autophagy-pyroptosis balance, while Nrf2 and FDX1 bridge ferroptosis-cuproptosis crosstalk. Therapeutic interventions targeting these mechanisms have shown promise in preclinical models. However, challenges persist, including the tissue-specific roles of gasdermin isoforms, off-target effects of pharmacological inhibitors, and transgenerational epigenetic impacts of environmental toxins. This review synthesizes current molecular insights into the cell death pathways implicated in male infertility, emphasizing their interplay and translational potential for restoring spermatogenic function.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Wei Chen
- Health Science Center, Hunan Normal University, Changsha 410013, China; (D.C.); (J.L.); (Z.P.); (R.F.); (C.C.); (F.L.); (Y.L.); (Y.S.); (C.L.)
| |
Collapse
|
73
|
Wang H, Yang Y, Zhang G, Yang G, Wang Y, Liu L, Du J. Roles of anoikis in hepatocellular carcinoma therapy and the assessment of anoikis-regulatory molecules as therapeutic targets. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04088-w. [PMID: 40183941 DOI: 10.1007/s00210-025-04088-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 03/21/2025] [Indexed: 04/05/2025]
Abstract
As the fourth leading cause of death from cancer and the sixth most common neoplasm in the world, hepatocellular carcinoma (HCC) is responsible for ninety percent of all primary liver cancers. There are four mechanisms that contribute to the spread of cancer: the separation of cells from the primary neoplasm, their survivability during metastasis, extravasation, and the development of secondary tumors at remote locations. In addition to its role in the development of a scaffold for cell adhesion, the extracellular matrix (ECM) also plays a role in the stimulation of signal transduction and the regulation of essential cellular mechanisms, including proliferation, migration, differentiation, and viability. The disruption of cell-ECM interactions and the ensuing separation of cells from the primary ECM trigger anoikis, a form of programmed cell death. One of the most effective factors in suppressing anoikis is ECM receptors from the integrin family. Cell migration, proliferation, and survival are primarily governed by the formation of physical connections with the cytoskeleton and the conveyance of signals between cells and the ECM via integrin receptors.
Collapse
Affiliation(s)
- Hongyu Wang
- Department of Abdominal Oncology, Jilin Cancer Hospital, Changchun, 130000, China
| | - Yawen Yang
- Department of Abdominal Oncology, Jilin Cancer Hospital, Changchun, 130000, China
| | - Gan Zhang
- Department of Abdominal Oncology, Jilin Cancer Hospital, Changchun, 130000, China
| | - Guang Yang
- Department of Abdominal Oncology, Jilin Cancer Hospital, Changchun, 130000, China
| | - Ying Wang
- Department of Abdominal Oncology, Jilin Cancer Hospital, Changchun, 130000, China
| | - Lu Liu
- Department of Abdominal Oncology, Jilin Cancer Hospital, Changchun, 130000, China
| | - Juan Du
- Department of Abdominal Oncology, Jilin Cancer Hospital, Changchun, 130000, China.
| |
Collapse
|
74
|
Osterli E, Park Y, Hu K, Kasof G, Wiederhold T, Liu C, Hu B. The role of autophagy in ischemic brain injury. AUTOPHAGY REPORTS 2025; 4:2486445. [PMID: 40395988 PMCID: PMC11980474 DOI: 10.1080/27694127.2025.2486445] [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] [Received: 08/20/2024] [Revised: 02/26/2025] [Accepted: 03/11/2025] [Indexed: 05/22/2025]
Abstract
Ischemic brain injury occurs in many clinical settings, including stroke, cardiac arrest, hypovolemic shock, cardiac surgery, cerebral edema, and cerebral vasospasm. Decades of work have revealed many important mechanisms related to ischemic brain injury. However, there remain significant gaps in the scientific knowledge to reconcile many ischemic brain injury events. Brain ischemia leads to protein misfolding and aggregation, and damages almost all types of subcellular organelles including mitochondria, endoplasmic reticulum, Golgi apparatus, lysosomes, etc. Irreparably damaged organelles and insoluble protein aggregates are normally removed by autophagy. The build-up of common autophagic components, such as LC3, p62, and ubiquitinated proteins, are generally observed in brain tissue samples in animal models of both global and focal brain ischemia, but the interpretation of the role of these autophagy-related changes in ischemic brain injury in the literature has been controversial. Many pathological events or mechanisms underlying dysfunctional autophagy after brain ischemia remain unknown. This review aims to provide an update of the current knowledge and future research directions regarding the critical role of dysfunctional autophagy in ischemic brain injury.
Collapse
Affiliation(s)
- Emily Osterli
- Departments of Emergency Medicine and Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Yujung Park
- Departments of Emergency Medicine and Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Kurt Hu
- Department of Medicine, Division of Pulmonary and Critical Care, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Gary Kasof
- Departments of Product Design and Strategy and Antibody Protein Technology, Cell Signaling Technology, Danvers, MA, USA
| | - Thorsten Wiederhold
- Departments of Product Design and Strategy and Antibody Protein Technology, Cell Signaling Technology, Danvers, MA, USA
| | - Chunli Liu
- Departments of Emergency Medicine and Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Bingren Hu
- Departments of Emergency Medicine and Neurosciences, University of California San Diego, La Jolla, CA, USA
- Department of Research, Veterans Affairs San Diego Healthcare System, La Jolla, CA, USA
| |
Collapse
|
75
|
Dravecka M, Mikkola I, Johansen T, Seternes OM, Mejlvang J. Low extracellular pH protects cancer cells from ammonia toxicity. Cell Death Discov 2025; 11:137. [PMID: 40180899 PMCID: PMC11968834 DOI: 10.1038/s41420-025-02440-w] [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: 12/09/2024] [Revised: 03/12/2025] [Accepted: 03/25/2025] [Indexed: 04/05/2025] Open
Abstract
Ammonia is a natural waste product of cellular metabolism which, through its lysosomotropic ability, can have detrimental effects on various cellular functions. Increased levels of ammonia were recently detected in the interstitial fluid of various tumours, substantiating that high ammonia concentrations are a pathophysiological condition in the tumour microenvironment, alongside hypoxia and acidosis. Since little is known about how cancer cells respond to elevated levels of ammonia in the tumour microenvironment, we investigated how a panel of cancer cell lines derived from solid tumours behaved when exposed to increasing concentrations of ammonia. We found that ammonia represses cell growth, induces genome instability, and inhibits lysosome-mediated proteolysis in a dose-dependent manner. Unexpectedly, we also found that small fluctuations in the pH of the extracellular environment, had a significant impact on the cytotoxic effects of ammonia. In summary, our data show that the balance of pH and ammonia within the interstitial fluids of cancerous tumours significantly impacts the behaviour and fate of cells residing in the tumour microenvironment.
Collapse
Affiliation(s)
- Maria Dravecka
- Cell Signalling and Targeted therapy, Department of Pharmacy, UiT The Arctic University of Norway, Tromsø, Norway
| | - Ingvild Mikkola
- Cell Signalling and Targeted therapy, Department of Pharmacy, UiT The Arctic University of Norway, Tromsø, Norway
| | - Terje Johansen
- Autophagy Research Group, Department of Medical Biology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Ole Morten Seternes
- Cell Signalling and Targeted therapy, Department of Pharmacy, UiT The Arctic University of Norway, Tromsø, Norway
| | - Jakob Mejlvang
- Cell Signalling and Targeted therapy, Department of Pharmacy, UiT The Arctic University of Norway, Tromsø, Norway.
| |
Collapse
|
76
|
Shojaei S, Barzegar Behrooz A, Cordani M, Aghaei M, Azarpira N, Klionsky DJ, Ghavami S. A non-fluorescent immunohistochemistry method for measuring autophagy flux using MAP1LC3/LC3 and SQSTM1 as core markers. FEBS Open Bio 2025. [PMID: 40181489 DOI: 10.1002/2211-5463.70014] [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: 06/27/2024] [Revised: 12/31/2024] [Accepted: 02/23/2025] [Indexed: 04/05/2025] Open
Abstract
Macroautophagy/autophagy is a crucial cellular process for degrading and recycling damaged proteins and organelles, playing a significant role in diseases such as cancer and neurodegeneration. Evaluating autophagy flux, which tracks autophagosome formation, maturation, and degradation, is essential for understanding disease mechanisms. Current fluorescence-based methods are resource-intensive, requiring advanced equipment and expertise, limiting their use in clinical laboratories. Here, we introduce a non-fluorescent immunohistochemistry (IHC) method using MAP1LC3/LC3 and SQSTM1 as core markers for autophagy flux assessment. LC3 levels reflect autophagosome formation, whereas SQSTM1 degradation and a decrease in the number of its puncta indicate active flux (i.e., lysosomal turnover). We optimized chromogenic detection using diaminobenzidine (DAB) staining and developed a scoring system based on puncta number and the percentage of stained cells. This accessible, cost-effective method enables reliable autophagy quantification using a standard light microscope, bridging the gap between experimental research and clinical diagnostics. Our protocol allows accurate autophagy evaluation in fixed tissues, offering practical applications in biomedical research and clinical pathology assessment.
Collapse
Affiliation(s)
- Shahla Shojaei
- Department of Human Anatomy and Cell Science, College of Medicine, University of Manitoba, Winnipeg, Canada
| | - Amir Barzegar Behrooz
- Department of Human Anatomy and Cell Science, College of Medicine, University of Manitoba, Winnipeg, Canada
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, Complutense University, Madrid, Spain
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, Spain
| | - Mahmoud Aghaei
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Negar Azarpira
- Shiraz Institute for Stem Cell & Regenerative Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Daniel J Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, College of Medicine, University of Manitoba, Winnipeg, Canada
- Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Canada
- Research Institute of Oncology and Hematology, Cancer Care Manitoba, University of Manitoba, Winnipeg, Canada
- Faculty of Medicine, Academy of Silesia, Katowice, Poland
| |
Collapse
|
77
|
Pfau DJ, Bryk R. High throughput screening assay for the identification of ATF4 and TFEB activating compounds. AUTOPHAGY REPORTS 2025; 4:2473765. [PMID: 40265045 PMCID: PMC11980509 DOI: 10.1080/27694127.2025.2473765] [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] [Received: 07/10/2024] [Revised: 01/16/2025] [Accepted: 01/16/2025] [Indexed: 04/24/2025]
Abstract
Macrophages act to defend against infection, but can fail to completely prevent bacterial replication and dissemination in an immunocompetent host. Recent studies have shown that activation of a host transcription factor, TFEB, a regulator of lysosomal biogenesis, could restrict intramacrophage replication of the human pathogen Mycobacterium tuberculosis and synergize with suboptimal levels of the antibiotic rifampin to reduce bacterial loads. Currently available small molecule TFEB activators lack selectivity and potency, but could be potentially useful in a variety of pathological conditions with suboptimal lysosomal activity. TFEB nuclear translocation and activation depend on its phosphorylation status which is controlled by multiple cellular pathways. We devised a whole cell, high throughput screening assay to identify small molecules that activate TFEB by establishing a stably transfected HEK293T reporter cell line for ATF4, a basic leucine zipper transcription factor induced by stress response and activated in parallel to TFEB. We optimized its use in vitro using compounds that target endoplasmic reticulum stress and intracellular calcium signaling. We report results from screening the commercially available LOPAC library and the Selleck Chemicals library modified to include only FDA-approved drugs and clinical research compounds. We identified twenty-one compounds across six clinical use categories that activate ATF4, and confirmed that two proteasome inhibitors promote TFEB activation. The results of this study provide an assay that could be used to screen for small molecules that activate ATF4 and TFEB and a potential list of compounds identified as activators of the ATF4 transcription factor in response to cellular stress.
Collapse
Affiliation(s)
- Daniel J. Pfau
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA
| | - Ruslana Bryk
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
78
|
Caldera L, Masci C, Cappozzo A, Forlani M, Antonelli B, Leoni O, Ieva F. Uncovering mortality patterns and hospital effects in COVID-19 heart failure patients: a novel multilevel logistic cluster-weighted modeling approach. Biometrics 2025; 81:ujaf046. [PMID: 40293213 DOI: 10.1093/biomtc/ujaf046] [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: 06/26/2024] [Revised: 02/21/2025] [Accepted: 04/02/2025] [Indexed: 04/30/2025]
Abstract
Evaluating hospital performance and its relationship to patients' characteristics is of utmost importance to ensure timely, effective, and optimal treatment. This is particularly relevant in areas and situations where the healthcare system must deal with an unexpected surge in hospitalizations, such as heart failure patients in the Lombardy Region of Italy during the COVID-19 pandemic. Motivated by this issue, the paper introduces a novel multilevel logistic cluster-weighted model for predicting 45-day mortality following hospitalization due to COVID-19. The methodology flexibly accommodates dependence patterns among continuous and dichotomous variables; effectively accounting for group-specific effects in distinct subgroups showing different attributes. A tailored classification expectation-maximization algorithm is developed for parameter estimation, and extensive simulation studies are conducted to evaluate its performance against competing models. The novel approach is applied to administrative data from the Lombardy Region, with the aim of profiling heart failure patients hospitalized for COVID-19 and investigating the hospital-level impact on their overall mortality. A scenario analysis demonstrates the model's efficacy in managing multiple sources of heterogeneity, thereby yielding promising results in aiding healthcare providers and policymakers in the identification of patient-specific treatment pathways.
Collapse
Affiliation(s)
- Luca Caldera
- MOX, Department of Mathematics, Politecnico di Milano, Via Bonardi 9, Milan 20133, Italy
| | - Chiara Masci
- Department of Economics, Management, and Quantitative Methods, University of Milan, Via Festa del Perdono 7, Milan 20122, Italy
| | - Andrea Cappozzo
- Department of Statistical Sciences, Università Cattolica del Sacro Cuore, Largo Gemelli 1, Milan 20123, Italy
| | - Marco Forlani
- Regione Lombardia, Informatica SPA, Piazza Città di Lombardia 1, Milan 20124, Italy
| | - Barbara Antonelli
- Regione Lombardia, Divisione Servizi per il Welfare Regionale, Piazza Città di Lombardia 1, Milan 20124, Italy
| | - Olivia Leoni
- U.O. Osservatorio Epidemiologico, DG Welfare, Regione Lombardia, Piazza Città di Lombardia 1, Milan 20124, Italy
| | - Francesca Ieva
- MOX, Department of Mathematics, Politecnico di Milano, Via Bonardi 9, Milan 20133, Italy
- Health Data Science Centre, Human Technopole, Viale Rita Levi-Montalcini 1, Milan 20157, Italy
| |
Collapse
|
79
|
Subramanian SP, Wojtkiewicz M, Yu F, Castro C, Schuette EN, Rodriguez-Paar J, Churko J, Renavikar P, Anderson D, Mahr C, Gundry RL. Integrated Multiomics Reveals Alterations in Paucimannose and Complex Type N-Glycans in Cardiac Tissue of Patients with COVID-19. Mol Cell Proteomics 2025; 24:100929. [PMID: 39988192 DOI: 10.1016/j.mcpro.2025.100929] [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: 10/07/2024] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025] Open
Abstract
Coronavirus infectious disease of 2019 (COVID-19) can lead to cardiac complications, yet the molecular mechanisms driving these effects remain unclear. Protein glycosylation is crucial for viral replication, immune response, and organ function and has been found to change in the lungs and liver of patients with COVID-19. However, how COVID-19 impacts cardiac protein glycosylation has not been defined. Our study combined single nuclei transcriptomics, mass spectrometry (MS)-based glycomics, and lectin-based tissue imaging to investigate alterations in N-glycosylation in the human heart post-COVID-19. We identified significant expression differences in glycogenes involved in N-glycan biosynthesis and MS analysis revealed a reduction in high mannose and isomers of paucimannose structures post-infection, with changes in paucimannose directly correlating with COVID-19 independent of comorbidities. Our observations suggest that COVID-19 primes cardiac tissues to alter the glycome at all levels, namely, metabolism, nucleotide sugar transport, and glycosyltransferase activity. Given the role of N-glycosylation in cardiac function, this study provides a basis for understanding the molecular events leading to cardiac damage post-COVID-19 and informing future therapeutic strategies to treat cardiac complications resulting from coronavirus infections.
Collapse
Affiliation(s)
- Sabarinath Peruvemba Subramanian
- CardiOmics Program, Center for Heart and Vascular Research, and Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, USA.
| | - Melinda Wojtkiewicz
- CardiOmics Program, Center for Heart and Vascular Research, and Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Fang Yu
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Chase Castro
- CardiOmics Program, Center for Heart and Vascular Research, and Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Erin N Schuette
- CardiOmics Program, Center for Heart and Vascular Research, and Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Jocelyn Rodriguez-Paar
- CardiOmics Program, Center for Heart and Vascular Research, and Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Jared Churko
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA
| | - Pranav Renavikar
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Daniel Anderson
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Claudius Mahr
- Institute for Advanced Cardiac Care, Medical City Healthcare, Dallas, Texas, USA
| | - Rebekah L Gundry
- CardiOmics Program, Center for Heart and Vascular Research, and Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, USA.
| |
Collapse
|
80
|
Kang S, Liu S, Dong X, Li H, Qian Y, Dai A, He W, Li X, Chen Q, Wang H, Ding PH. USP4 depletion-driven RAB7A ubiquitylation impairs autophagosome-lysosome fusion and aggravates periodontitis. Autophagy 2025; 21:771-788. [PMID: 39663592 PMCID: PMC11925113 DOI: 10.1080/15548627.2024.2429371] [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: 04/03/2024] [Revised: 11/02/2024] [Accepted: 11/11/2024] [Indexed: 12/13/2024] Open
Abstract
Periodontitis, a prevalent and chronic inflammatory disease, is intricately linked with macroautophagy/autophagy, which has a dual role in maintaining periodontal homeostasis. Despite its importance, the precise interplay between autophagy and periodontitis pathogenesis remains to be fully elucidated. In this study, our investigation revealed that the ubiquitination of RAB7A, mediated by reduced levels of the deubiquitinating enzyme USP4 (ubiquitin specific peptidase 4), disrupts normal lysosomal trafficking and autophagosome-lysosome fusion, thereby contributing significantly to periodontitis progression. Specifically, through genomic and histological analysis of clinical gingival samples, we observed a decreased RAB7A expression and impaired autophagic activity in periodontitis. This was further substantiated through experimental periodontitis mice, where RAB7A inactivation was shown to directly affect autophagy efficiency and drive periodontitis progression. Next, we explored the function of active RAB7A to promote lysosomal trafficking dynamics and autophagosome-lysosome fusion, which was inhibited by RAB7A ubiquitination in macrophages stimulated by Porphyromonas gingivalis (P. g.), one of the keystone pathogens of periodontitis. Last, by proteomics analysis, we revealed that the ubiquitination of RAB7A was mediated by USP4 and validated that upregulation of USP4 could attenuate periodontitis in vivo. In conclusion, these findings highlight the interaction between USP4 and RAB7A as a promising target for therapeutic intervention in managing periodontal diseases.Abbreviation: 3-MA: 3-methyladenine; Baf A1:bafilomycin A1; BECN1: beclin 1, autophagy related; CEJ-ABC: cementoenamel junctionto alveolar bone crest; IL1B/IL-1β: interleukin 1 beta; KD:knockdown; LPS: lipopolysaccharide; MOI: multiplicity of infection;OE: overexpression; P.g.: Porphyromonasgingivalis; RILP: Rabinteracting lysosomal protein; ScRNA-seq: single-cell RNA sequencing; SQSTM1/p62: sequestosome 1; S.s.: Streptococcus sanguinis; USP4:ubiquitin specific peptidase 4.
Collapse
Affiliation(s)
- Sen Kang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
| | - Shuxin Liu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
| | - Xian Dong
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
| | - Haoyu Li
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuanyi Qian
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
| | - Anna Dai
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
| | - Wentao He
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaojun Li
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
| | - Qianming Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
| | - Huiming Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
| | - Pei-Hui Ding
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
81
|
Guo L, Wang N, Chen J, Zhang R, Li D, Yang L. Cellular senescence and glaucoma. Exp Gerontol 2025; 202:112718. [PMID: 39983803 DOI: 10.1016/j.exger.2025.112718] [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: 11/13/2024] [Revised: 01/06/2025] [Accepted: 02/18/2025] [Indexed: 02/23/2025]
Abstract
Cellular senescence, a characteristic feature of the aging process, is induced by diverse stressors. In recent years, glaucoma has emerged as a blinding ocular disease intricately linked to cellular senescence. The principal pathways implicated are oxidative stress, mitochondrial dysfunction, DNA damage, autophagy impairment, and the secretion of various senescence- associated secretory phenotype factors. Research on glaucoma-associated cellular senescence predominantly centers around the increased resistance of the aqueous humor outflow pathway, which is attributed to the senescence of the trabecular meshwork and Schlemm's canal. Additionally, it focuses on the mechanisms underlying retinal ganglion cell senescence in glaucoma and the corresponding intervention measures. Given that cell senescence represents an irreversible phase preceding cell death, an in-depth investigation into its mechanisms in the pathogenesis and progression of glaucoma, particularly by specifically blocking the signal transduction of cell senescence, holds the potential to decrease the outflow resistance of aqueous humor. This, in turn, could provide a novel avenue for safeguarding the optic nerve in glaucoma.
Collapse
Affiliation(s)
- Liang Guo
- The Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Na Wang
- The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Jing Chen
- The Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Rui Zhang
- The Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Dan Li
- The Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Lu Yang
- The Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, China.
| |
Collapse
|
82
|
Masson CD, Findlay-Greene F, Sousa FH, Henderson P, Fraser JA, Barlow PG, Stevens C. Characterisation of autophagy induction by the thiopurine drugs azathioprine, mercaptopurine and thioguanine in THP-1 macrophages. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:4467-4478. [PMID: 39485532 PMCID: PMC11978722 DOI: 10.1007/s00210-024-03563-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 10/22/2024] [Indexed: 11/03/2024]
Abstract
Activating autophagy may be therapeutically beneficial, and we have previously shown that azathioprine (AZA), an immunomodulatory drug, induces autophagy. Here, we evaluated the induction of autophagy by the thiopurines AZA, mercaptopurine (6-MP) and thioguanine (6-TG) in THP-1 macrophages and investigated the mechanism of action in the context of this cellular process. The cytotoxicity of thiopurines was evaluated using an LDH assay. Induction of endogenous LC3 by thiopurines was evaluated using immunostaining. To confirm autophagy activation by thiopurines, a GFP-RFP-LC3 reporter plasmid was used to monitor the maturation of autophagosomes to autolysosomes. Induction of apoptosis by thiopurines was evaluated using Annexin V/PI staining, and ER stress was assessed via RT‒PCR analysis of XBP1 splicing. To gain insight into the mechanism of action of thiopurines, mTORC1 activity and eIF2α-S51 phosphorylation were evaluated by immunoblotting. Thiopurines were not cytotoxic to cells and induced strong time- and concentration-dependent autophagy. Thiopurines activate autophagy with complete progression through the pathway. Induction of autophagy by thiopurines occurred independently of apoptosis and ER stress. Immunoblotting revealed that AZA inhibited mTORC1 activity, and AZA and 6-TG increased eIF2α-S51 phosphorylation. In contrast, 6-MP had a minor effect on either signalling pathway. Thiopurines are strong inducers of autophagy, and autophagy induction should be considered among the mechanisms responsible for patient response to thiopurines.
Collapse
Affiliation(s)
- Connan D Masson
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Sighthill Court, Edinburgh, EH11 4BN, UK
| | - Fern Findlay-Greene
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Sighthill Court, Edinburgh, EH11 4BN, UK
| | - Filipa Henderson Sousa
- Centre for Discovery Brain Sciences and UK Dementia Research Institute, The University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - Paul Henderson
- Child Life and Health, University of Edinburgh, Edinburgh, EH16 4TJ, UK
- Department of Paediatric Gastroenterology and Nutrition, Royal Hospital for Children and Young People, Edinburgh, EH16 4TJ, UK
| | - Jennifer A Fraser
- Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Easter Bush Campus, Edinburgh, EH25 9RG, UK
| | - Peter G Barlow
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Sighthill Court, Edinburgh, EH11 4BN, UK
| | - Craig Stevens
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Sighthill Court, Edinburgh, EH11 4BN, UK.
| |
Collapse
|
83
|
Li Y, Shi J, Liu C, Ma D, Meng L, Zhang Z, Jia H. Ciprofol reduces postoperative glioma recurrence by promoting MAPK11-PML phosphorylation: insights from transcriptomic and proteomic analysis. J Neurooncol 2025; 172:361-376. [PMID: 40019711 DOI: 10.1007/s11060-024-04906-w] [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: 10/22/2024] [Accepted: 12/03/2024] [Indexed: 03/01/2025]
Abstract
BACKGROUND Glioma, the most common malignant tumor of the central nervous system, has a high postoperative recurrence rate. While Ciprofol is widely used as an anesthetic, its therapeutic potential in glioma treatment remains largely unexplored. METHODS Glioma T98G cells were treated with varying concentrations of Ciprofol to assess proliferation, invasion, migration, and apoptosis via CCK-8, Transwell, and flow cytometry assays. Proteomic, phosphoproteomic, and transcriptomic analyses were performed to identify molecular targets and pathways. Molecular docking evaluated the binding of Ciprofol to key kinases, and silencing experiments validated their roles. In vivo, glioma mouse models were used to assess postoperative recurrence via tumor size, fluorescence imaging, and histological analysis. RESULTS Ciprofol inhibited glioma cell proliferation, invasion, and migration while promoting apoptosis. Proteomic analyses identified MAPK11 and PML as key mediators of Ciprofol's effects. Silencing MAPK11 impaired these effects, while in vivo experiments showed reduced postoperative recurrence via MAPK11-PML phosphorylation. CONCLUSION Ciprofol reduces postoperative glioma recurrence by promoting MAPK11-PML phosphorylation, providing novel molecular targets for glioma treatment and suggesting its therapeutic potential beyond anesthesia.
Collapse
Affiliation(s)
- Yanli Li
- Department of Anesthesiology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Jingpu Shi
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, NO.12, JianKang Road, Shijiazhuang, 050011, Hebei, China
| | - Chao Liu
- Department of Anesthesiology and Intensive Care, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Dongyang Ma
- Department of Anesthesiology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Lijiang Meng
- Department of Anesthesiology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Zhiqiang Zhang
- Department of Anesthesiology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Huiqun Jia
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, NO.12, JianKang Road, Shijiazhuang, 050011, Hebei, China.
| |
Collapse
|
84
|
Walter S, Häseli SP, Baumgarten P, Deubel S, Jung T, Höhn A, Ott C, Grune T. Oxidized protein aggregate lipofuscin impairs cardiomyocyte contractility via late-stage autophagy inhibition. Redox Biol 2025; 81:103559. [PMID: 40068328 PMCID: PMC11938141 DOI: 10.1016/j.redox.2025.103559] [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: 12/10/2024] [Revised: 02/10/2025] [Accepted: 02/18/2025] [Indexed: 03/22/2025] Open
Abstract
Aging of the heart is accompanied by impairment of cardiac structure and function. At molecular level, autophagy plays a crucial role in preserving cardiac health. Autophagy maintains cellular homeostasis by facilitating balanced degradation of cytoplasmic components including organelles and misfolded or aggregated proteins. The age-related decline in autophagy favors an accumulation of protein aggregates such as lipofuscin particularly in the heart, which is composed primarily of non-proliferating cells. Therefore, this study investigates whether lipofuscin accumulation contributes to age-related functional decline of primary adult cardiomyocytes isolated from C57BL/6J mice and examines the role of autophagic flux in mediating these effects. Results showed an age-associated reduction in cardiomyocyte contraction amplitude and an increase in autofluorescence, indicating the accumulation of lipofuscin with age. In vitro treatment of adult primary cardiomyocytes with artificial lipofuscin increased autofluorescence and decreased both contraction amplitude and cellular autophagic flux. Induction of autophagy with rapamycin mitigated contractile dysfunction in lipofuscin-treated cardiomyocytes, whereas inhibition of autophagic flux revealed stage-dependent effects. Late-stage autophagy inhibition using chloroquine or concanamycin A reduced cardiomyocyte contraction amplitude, whereas early-stage autophagy inhibition via 3-methyladenine did not affect contraction within 24 h. In conclusion, our results indicate that lipofuscin directly impairs cardiomyocyte function by diminishing late-stage autophagic flux. These findings highlight the essential role of the autophagy-lysosomal system in preserving age-related loss of cardiomyocyte function caused by accumulating protein aggregates.
Collapse
Affiliation(s)
- Sophia Walter
- Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany; TraceAge-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly, Potsdam-Berlin-Jena-Wuppertal, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Steffen P Häseli
- Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Patricia Baumgarten
- Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Stefanie Deubel
- Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany
| | - Tobias Jung
- Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany
| | - Annika Höhn
- Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Christiane Ott
- Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany; TraceAge-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly, Potsdam-Berlin-Jena-Wuppertal, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany; Cardiac Aging and Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany.
| | - Tilman Grune
- Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany; TraceAge-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly, Potsdam-Berlin-Jena-Wuppertal, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany; Institute of Nutritional Science, Department of Food Chemistry, University of Potsdam, Potsdam, 14469, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| |
Collapse
|
85
|
Fu D, Li Z, Feng H, Fan F, Zhang W, He L. Chaperone mediated autophagy modulates microglia polarization and inflammation via LAMP2A in ischemia induced spinal cord injury. Toxicol Res (Camb) 2025; 14:tfaf061. [PMID: 40309223 PMCID: PMC12038812 DOI: 10.1093/toxres/tfaf061] [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: 01/27/2025] [Revised: 03/17/2025] [Accepted: 04/14/2025] [Indexed: 05/02/2025] Open
Abstract
Spinal cord injury (SCI)-induced ischemic delayed paralysis is one of the most serious side effects of aneurysms surgeries. Recent studies prove that the activation of autophagy, including macroautophagy and micro-autophagy pathways, occur during SCI-induced brain neuron damage. However, the role of chaperone mediated autophagy (CMA) during SCI remains to be unveiled. In the present work, rat model of delayed paralysis after aneurysms operation and adenovrius induced LAMP2A knockdown in microglia cells were applied in the present work to investigate the involvement of LAMP2A-mediated CMA in the aneurysm operation related SCI and delayed paralysis. The results showed that LAMP2A was upregulated in the SCI procedure, and contributed to neuron death and pro-inflammation perturbation via inducing iNOS+ polarization in microgila. We additionally observed that knockdown of LAMP2A resulted in the shift of microglia from iNOS+ to ARG1+ phenotype, as well as alleviated neuron damage during SCI. Furthermore, the analysis of BBB score, the result of immunohistological staining, and protein detection confirmed the activation of LAMP2A-mediated CMA activation and its interaction with NF-κB signaling, which leads to neuron death and motor function loss. These results prove that LAMP2A-mediated CMA contributes to the upregulation of pro-inflammatory cytokines and results in cell death in neurons during ischemic delayed paralysis via activating NF-κB signaling. Inhibition of LAMP2A promotes neurons survival during ischemic delayed paralysis.
Collapse
Affiliation(s)
- Dan Fu
- Department of Pediatrics, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou City, Guangdong Province 510900, China
| | - Ziyou Li
- Department of Anesthesiology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou City, Guangdong Province 510900, China
| | - Huafeng Feng
- Department of Anesthesiology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou City, Guangdong Province 510900, China
| | - Fangling Fan
- Department of Anesthesiology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou City, Guangdong Province 510900, China
| | - Wang Zhang
- Department of Anesthesiology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou City, Guangdong Province 510900, China
| | - Liang He
- Department of Anesthesiology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou City, Guangdong Province 510900, China
| |
Collapse
|
86
|
Zhao X, Zhang X, Wu L, Liu X, Pan Y, Lv T, Xu M, Yang K, Wang X. WTAP suppresses STAT3 via m6A methylation to regulate autophagy and inflammation in central nervous system injury. Neurobiol Dis 2025; 207:106811. [PMID: 39855477 DOI: 10.1016/j.nbd.2025.106811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 01/15/2025] [Accepted: 01/21/2025] [Indexed: 01/27/2025] Open
Abstract
Central nervous system (CNS) repair after injury is a challenging process limited by inflammation and neuronal apoptosis. This study identifies Wilms' tumor 1-associating protein (WTAP) as a pivotal regulator of neuronal protection and repair through m6A methylation of STAT3 mRNA. By employing spinal cord injury (SCI) as a representative model of CNS injury, transcriptomic analyses reveal WTAP as a key mediator of pathways related to neuronal autophagy and inflammation regulation. WTAP enhances neuronal autophagy by suppressing STAT3 expression and activity, which inhibits the NLRP3 inflammatory pathway. Functional studies demonstrate that WTAP knockdown exacerbates neuronal apoptosis, whereas overexpression improves cell viability, autophagy, and motor recovery. In vivo, WTAP promotes SCI repair via m6A-mediated suppression of STAT3 and regulation of the NLRP3 signaling pathway, highlighting its therapeutic potential for CNS injury repair.
Collapse
Affiliation(s)
- Xiaoyong Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China; Department of Neurosurgery, The Fifth Hospital of Guangzhou Medical University, Guangzhou 510632, China
| | - Xiaoli Zhang
- Department of Obstetrics and Gynecology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510632, China.
| | - Liangzhi Wu
- Department of Obstetrics and Gynecology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510632, China
| | - Xiaohe Liu
- Department of Neurosurgery, The Fifth Hospital of Guangzhou Medical University, Guangzhou 510632, China
| | - Yongquan Pan
- Department of Neurosurgery, Huadu District People's Hospital of Guangzhou, Guangzhou 510800, China
| | - Taiquan Lv
- Department of Neurosurgery, Huadu District People's Hospital of Guangzhou, Guangzhou 510800, China
| | - Mingyang Xu
- Department of Neurosurgery, The Fifth Hospital of Guangzhou Medical University, Guangzhou 510632, China
| | - Kongbin Yang
- Department of Neurosurgery, The Fifth Hospital of Guangzhou Medical University, Guangzhou 510632, China.
| | - Xiangyu Wang
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China.
| |
Collapse
|
87
|
Liu XW, Huang SS, Xu P, Xu HW, Wang DK, Wang SJ. Transcription factor EP300 targets SIRT5 to promote autophagy of nucleus pulposus cells and attenuate intervertebral disc degeneration. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119933. [PMID: 40096894 DOI: 10.1016/j.bbamcr.2025.119933] [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: 11/17/2024] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND Intervertebral disc degeneration (IVDD) is a prevalent spinal ailment and the leading cause of chronic low back pain. Understanding the exact pathogenesis of IVDD and developing targeted molecular drugs will be important in the future. Autophagy plays a key role in the metabolic processes and in the quality control of proteins in IVDD. However, the role of autophagy in the senescence of nucleus pulposus cell (NPC), the primary cells in the intervertebral disc responsible for maintaining the disc's structure and function, is not yet clear. METHODS Gene expression profiling data of human disc tissue were obtained from the Gene Expression Omnibus GSE15227, GSE23130, and GSE70362 datasets. Autophagy-related differentially expressed genes were identified from the Molecular Signatures Database (MSigDB) database. Weighted gene co-expression network analysis (WGCNA), receiver operating characteristic (ROC) curves, and least absolute shrinkage and selection operator (LASSO) regression identified an autophagy-related hub gene that encodes the E1A binding protein EP300 transcription factor in IVDD samples. Potential downstream target genes of EP300 were identified by bioinformatics analysis. The analysis identified sirtuin 5 (SIRT5) as a potential downstream target of EP300. Chromatin immunoprecipitation (ChIP)-qPCR, small interfering RNA (siRNA), and luciferase reporter gene assays were used to verify the interaction of EP300 and SIRT5 in vitro. For in vivo experiments, SIRT5 knockout mice and SIRT5-overexpressing adeno-associated virus serotype 5 (AAV5) were constructed to verify the effect of the EP300-SIRT5 signal axis on the progression of IVDD. RESULTS EP300 expression was reduced in the IVDD samples compared with its expression in healthy disc tissue samples. The reduced EP300 expression inhibited the occurrence of autophagy, which promoted NPC senescence. ChIP-qPCR and luciferase reporter gene assays showed that EP300 promoted SIRT5 expression by direct binding to its promoter. Activation of EP300 expression increased SIRT5 expression and significantly improved autophagy for inhibition of NPC senescence. In vivo experiments confirmed that knockdown of EP300 promoted NPC senescence and led to an exacerbation of IVDD, which was reversed by SIRT5 overexpression. CONCLUSION Our results provide the first evidence for the importance of EP300 and SIRT5 interactions in promoting IVDD development by inhibiting autophagy during IVDD. The EP300-SIRT5 signaling axis was identified as a promising target for therapy of IVDD based on autophagy genes.
Collapse
Affiliation(s)
- Xiao-Wei Liu
- Department of Spinal Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Shan-Shan Huang
- Department of Geriatric Neurology of Hua Shan Hospital, National Clinical Research Center for Aging and Medicine, Fudan University, Shanghai, China
| | - Pei Xu
- Department of Neurosurgery, The Central Hospital Affiliated to Shaoxing University, Shaoxing, Zhejiang Province, China
| | - Hao-Wei Xu
- Department of Spinal Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Dian-Kai Wang
- Department of Spinal Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Shan-Jin Wang
- Department of Spinal Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
88
|
Ye L, Wang L, Kuang G, Zhang Z, Peng Q, He M, Fan J. IL-27 aggravates acute hepatic injury by promoting macrophage M1 polarization to induce Caspase-11 mediated Pyroptosis in vitro and in vivo. Cytokine 2025; 188:156881. [PMID: 39913960 DOI: 10.1016/j.cyto.2025.156881] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 01/24/2025] [Accepted: 02/01/2025] [Indexed: 03/01/2025]
Abstract
OBJECTIVES Our aim was to explore the IL-27 effect in sepsis (SP)-related acute hepatic injury (AHI) as well as its possible mechanism. MATERIALS AND METHODS Herein, we utilized both wild-type (WT) and IL-27 receptor (WSX-1)-deficient (IL-27R-/-) mice alongside RAW264.7 cells. Our study established an SP-associated AHI model through the intraperitoneal injections of lipopolysaccharide (LPS) + D-galactosamine (D-G). For examining the IL-27 impact on AHI, mice serum and liver tissue samples were gathered. Inflammatory factor levels in the liver and serum were detected using ELISA and immunohistochemistry. Immunofluorescence and Western blot techniques were employed for the detection of protein expression associated with polarization and pyroptosis in the liver, including iNOS, ARG-1, caspase-11, RAGE, and GSDMD. To further verify the IL-27 effects on macrophage polarization and pyroptosis and explore possible mechanisms involved, we used LPS-triggered RAW264.7 macrophages to assess AMPK/SIRT1 expression after IL-27 intervention. This study utilized Compound C (CC) to block the AMPK/SIRT1 pathway. The inflammatory response level and protein expression related to macrophage polarization and pyroptosis were measured again to reveal IL-27 implication in AHI and determine whether its role is associated with the AMPK/SIRT1 pathway. RESULTS The results revealed that IL-27 exacerbated systemic inflammation and liver damage in AHI mice by promoting M1 macrophage polarization, thereby increasing pro-inflammatory phenotype macrophages (M1). This further exacerbated the inflammatory response and pyroptosis in vivo and in vitro. Additionally, IL-27 down-regulated p-AMPK and SIRT1 protein expression while overexpressing macrophage inflammatory mediators including IL-1β/6 and TNFα. Furthermore, IL-27 promoted increased RAGE and caspase-11 protein expression, aggravating macrophage pyroptosis. Employing CC to block the AMPK pathway further aggravated M1 macrophage polarization and pyroptosis in vitro and in vivo, ultimately worsening liver injury. CONCLUSIONS Here, IL-27 aggravates AHI by promoting macrophage M1 polarization to induce caspase-11-mediated pyroptosis in vitro and in vivo, which may be linked to the AMPK/SIRT1 signaling pathway.
Collapse
Affiliation(s)
- Lin Ye
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, No.1, Youyi Road, Yuzhong District, Chongqing City 400016, China; The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical Universit, Chongqing City, China
| | - Liuyang Wang
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, No.1, Youyi Road, Yuzhong District, Chongqing City 400016, China; The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical Universit, Chongqing City, China
| | - Gang Kuang
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical Universit, Chongqing City, China; Department of Critical Care Medicine, Affiliated Dazu's Hospital of Chongqing Medical University, No. 1073, The second Ring South Road, Tangxiang Street, DaZu District, Chongqing City 402360, China
| | - Zhijiao Zhang
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, No.1, Youyi Road, Yuzhong District, Chongqing City 400016, China; The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical Universit, Chongqing City, China
| | - Qiaozhi Peng
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, No.1, Youyi Road, Yuzhong District, Chongqing City 400016, China; The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical Universit, Chongqing City, China
| | - Miao He
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Jing Fan
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, No.1, Youyi Road, Yuzhong District, Chongqing City 400016, China.
| |
Collapse
|
89
|
Marinković M, Rožić A, Polančec D, Novak I. Cost-effective and simple flow cytometry quantification of receptor-mediated autophagy using fluorescent tagging. FEBS Open Bio 2025; 15:587-598. [PMID: 39716041 PMCID: PMC11961372 DOI: 10.1002/2211-5463.13958] [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: 08/06/2024] [Revised: 11/20/2024] [Accepted: 12/13/2024] [Indexed: 12/25/2024] Open
Abstract
Mitophagy, a selective clearance of damaged or superfluous mitochondria via autophagy machinery and lysosomal degradation, is an evolutionarily conserved process essential for various physiological functions, including cellular differentiation and immune responses. Defects in mitophagy are implicated in numerous human diseases, such as neurodegenerative disorders, cancer, and metabolic conditions. Despite significant advancements in mitophagy research over recent decades, novel and robust methodologies are necessary to elucidate its molecular mechanisms comprehensively. In this study, we present a detailed protocol for quantitatively assessing mitophagy through flow cytometry using a mitochondria-targeted fluorescent mitophagy receptor, GFP-BNIP3L/NIX. This method offers a rapid alternative to conventional microscopy or immunoblotting techniques for analyzing mitophagy activity. Additionally, this approach can theoretically be adapted to utilize any fluorescent-tagged selective autophagy receptor, enabling the direct and rapid analysis of various types of receptor-mediated selective autophagy.
Collapse
Affiliation(s)
| | - Ana Rožić
- School of MedicineUniversity of SplitCroatia
| | | | - Ivana Novak
- School of MedicineUniversity of SplitCroatia
| |
Collapse
|
90
|
Guo Y, Zhang Q, Zhang B, Pan T, Ronan EA, Huffman A, He Y, Inoki K, Liu J, Xu XS. Dietary cinnamon promotes longevity and extends healthspan via mTORC1 and autophagy signaling. Aging Cell 2025; 24:e14448. [PMID: 39760475 PMCID: PMC11984692 DOI: 10.1111/acel.14448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/12/2024] [Accepted: 12/04/2024] [Indexed: 01/07/2025] Open
Abstract
Cinnamon, renowned for its aromatic flavor, represents one of the most widely used spices worldwide. Cinnamon is also considered beneficial to human health with therapeutic potential for treating various diseases, ranging from diabetes and cancer to neurodegenerative diseases. However, the mechanisms underlying cinnamon's health benefits remain elusive. It is also unclear whether cinnamon has any role in aging. Using C. elegans as a model, here we show that feeding worms cinnamaldehyde (CA), the active ingredient in cinnamon oil, prolongs longevity. CA also promotes stress resistance and reduces β-Amyloid toxicity in a C. elegans model of Alzheimer's disease. Mechanistically, CA exerts its beneficial effects through mTORC1 and autophagy signaling. Interestingly, CA promotes longevity by inducing a dietary restriction-like state without affecting food intake, suggesting CA as a dietary restriction mimetic. In human cells, CA exerts a similar effect on mTORC1 and autophagy signaling, suggesting a conserved mechanism. Our results demonstrate that dietary cinnamon promotes both lifespan and healthspan and does so by regulating mTORC1 and autophagy signaling.
Collapse
Affiliation(s)
- Yuling Guo
- College of Life Science and Technology, Key Laboratory of Molecular Biophysics of MOEHuazhong University of Science and TechnologyWuhanHubeiChina
- Life Sciences Institute, University of MichiganAnn ArborMichiganUSA
- Department of Molecular and Integrative PhysiologyUniversity of MichiganAnn ArborMichiganUSA
| | - Qing Zhang
- College of Life Science and Technology, Key Laboratory of Molecular Biophysics of MOEHuazhong University of Science and TechnologyWuhanHubeiChina
| | - Bi Zhang
- College of Life Science and Technology, Key Laboratory of Molecular Biophysics of MOEHuazhong University of Science and TechnologyWuhanHubeiChina
| | - Tong Pan
- Life Sciences Institute, University of MichiganAnn ArborMichiganUSA
- Department of Molecular and Integrative PhysiologyUniversity of MichiganAnn ArborMichiganUSA
| | - Elizabeth A. Ronan
- Life Sciences Institute, University of MichiganAnn ArborMichiganUSA
- Department of Molecular and Integrative PhysiologyUniversity of MichiganAnn ArborMichiganUSA
| | - Anthony Huffman
- Department of Computational Medicine and BioinformaticsUniversity of MichiganAnn ArborMichiganUSA
| | - Yongqun He
- Department of Computational Medicine and BioinformaticsUniversity of MichiganAnn ArborMichiganUSA
- Unit for Laboratory Animal MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Ken Inoki
- Life Sciences Institute, University of MichiganAnn ArborMichiganUSA
- Department of Molecular and Integrative PhysiologyUniversity of MichiganAnn ArborMichiganUSA
| | - Jianfeng Liu
- College of Life Science and Technology, Key Laboratory of Molecular Biophysics of MOEHuazhong University of Science and TechnologyWuhanHubeiChina
- Bioland LaboratoryGuangzhou Regenerative Medicine and Health Guangdong LaboratoryGuangzhouChina
| | - X.Z. Shawn Xu
- Life Sciences Institute, University of MichiganAnn ArborMichiganUSA
- Department of Molecular and Integrative PhysiologyUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
91
|
Li S, Zhou X, Duan Q, Niu S, Li P, Feng Y, Zhang Y, Xu X, Gong SP, Cao H. Autophagy and Its Association with Macrophages in Clonal Hematopoiesis Leading to Atherosclerosis. Int J Mol Sci 2025; 26:3252. [PMID: 40244103 PMCID: PMC11989900 DOI: 10.3390/ijms26073252] [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: 01/27/2025] [Revised: 03/04/2025] [Accepted: 03/17/2025] [Indexed: 04/18/2025] Open
Abstract
Atherosclerosis, a chronic inflammatory disease characterized by lipid accumulation and immune cell infiltration, is linked to plaque formation and cardiovascular events. While traditionally associated with lipid metabolism and endothelial dysfunction, recent research highlights the roles of autophagy and clonal hematopoiesis (CH) in its pathogenesis. Autophagy, a cellular process crucial for degrading damaged components, regulates macrophage homeostasis and inflammation, both of which are pivotal in atherosclerosis. In macrophages, autophagy influences lipid metabolism, cytokine regulation, and oxidative stress, helping to prevent plaque instability. Defective autophagy exacerbates inflammation, impairs cholesterol efflux, and accelerates disease progression. Additionally, autophagic processes in endothelial cells and smooth muscle cells further contribute to atherosclerotic pathology. Recent studies also emphasize the interplay between autophagy and CH, wherein somatic mutations in genes like TET2, JAK2, and DNMT3A drive immune cell expansion and enhance inflammatory responses in atherosclerotic plaques. These mutations modify macrophage function, intensifying the inflammatory environment and accelerating atherosclerosis. Chaperone-mediated autophagy (CMA), a selective form of autophagy, also plays a critical role in regulating macrophage inflammation by degrading pro-inflammatory cytokines and oxidized low-density lipoprotein (ox-LDL). Impaired CMA activity leads to the accumulation of these substrates, activating the NLRP3 inflammasome and worsening inflammation. Preclinical studies suggest that pharmacologically activating CMA may mitigate atherosclerosis progression. In animal models, reduced CMA activity accelerates plaque instability and increases inflammation. This review highlights the importance of autophagic regulation in macrophages, focusing on its role in inflammation, plaque formation, and the contributions of CH. Building upon current advances, we propose a hypothesis in which autophagy, programmed cell death, and clonal hematopoiesis form a critical intrinsic axis that modulates the fundamental functions of macrophages, playing a complex role in the development of atherosclerosis. Understanding these mechanisms offers potential therapeutic strategies targeting autophagy and inflammation to reduce the burden of atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Shuanhu Li
- Key Laboratory of Pharmacodynamics and Material Basis of Chinese Medicine of Shaanxi Administration of Traditional Chinese Medicine, Engineering Research Center of Brain Health Industry of Chinese Medicine, Pharmacology of Chinese Medicine, Shaanxi University of Chinese Medicine, University Government Committee of Shaanxi Province, Xianyang 712046, China;
| | - Xin Zhou
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi’an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi’an Medical University, Xi’an 710021, China; (S.N.); (P.L.); (Y.F.); (Y.Z.); (S.-P.G.)
| | - Qinchun Duan
- Laboratory of Cell Biology, Genetics and Developmental Biology, College of Life Sciences, Shaanxi Normal University, Xi’an 710062, China; (Q.D.); or (X.X.)
| | - Shukun Niu
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi’an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi’an Medical University, Xi’an 710021, China; (S.N.); (P.L.); (Y.F.); (Y.Z.); (S.-P.G.)
| | - Pengquan Li
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi’an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi’an Medical University, Xi’an 710021, China; (S.N.); (P.L.); (Y.F.); (Y.Z.); (S.-P.G.)
| | - Yihan Feng
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi’an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi’an Medical University, Xi’an 710021, China; (S.N.); (P.L.); (Y.F.); (Y.Z.); (S.-P.G.)
| | - Ye Zhang
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi’an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi’an Medical University, Xi’an 710021, China; (S.N.); (P.L.); (Y.F.); (Y.Z.); (S.-P.G.)
| | - Xuehong Xu
- Laboratory of Cell Biology, Genetics and Developmental Biology, College of Life Sciences, Shaanxi Normal University, Xi’an 710062, China; (Q.D.); or (X.X.)
| | - Shou-Ping Gong
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi’an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi’an Medical University, Xi’an 710021, China; (S.N.); (P.L.); (Y.F.); (Y.Z.); (S.-P.G.)
| | - Huiling Cao
- Key Laboratory of Pharmacodynamics and Material Basis of Chinese Medicine of Shaanxi Administration of Traditional Chinese Medicine, Engineering Research Center of Brain Health Industry of Chinese Medicine, Pharmacology of Chinese Medicine, Shaanxi University of Chinese Medicine, University Government Committee of Shaanxi Province, Xianyang 712046, China;
- Xi’an Key Laboratory of Basic and Translation of Cardiovascular Metabolic Disease, Xi’an Key Laboratory of Autoimmune Rheumatic Disease, College of Pharmacy, Xi’an Medical University, Xi’an 710021, China; (S.N.); (P.L.); (Y.F.); (Y.Z.); (S.-P.G.)
| |
Collapse
|
92
|
Dagher DM, Zaghloul MS, Suddek GM. Modulation of AMPK/mTOR Autophagic Pathway Using Dapagliflozin Protects Against Cadmium-Induced Testicular and Renal Injury in Rats. J Biochem Mol Toxicol 2025; 39:e70257. [PMID: 40233265 DOI: 10.1002/jbt.70257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 02/03/2025] [Accepted: 03/31/2025] [Indexed: 04/17/2025]
Abstract
Cadmium is a widely distributed heavy metal found in the environment that poses serious hazards to human health. Dapagliflozin (DAPA), a sodium-glucose co-transporter 2 (SGLT-2) inhibitor, exhibited antioxidant, antiapoptotic, and anti-inflammatory properties. Our data assessed the effect of DAPA against Cd-triggered renal and testicular impairment in rats, as well as the underlying mechanisms. Cd (30 mg/kg) and DAPA (5 and 10 mg/kg) were administrated by oral gavage to rats and continued for 21 days. DAPA attenuated Cd-triggered renal and testicular injury as shown by diminishing serum creatinine, BUN, and urinary total protein concentration in addition to increasing creatinine clearance, urinary creatinine, and serum testosterone. Moreover, it diminished renal and testicular histopathological alterations induced by Cd. DAPA stimulated the impaired autophagy flux as seen by significantly elevating the p-AMPK/total AMPK, decreasing p-mTOR/total mTOR ratios, and diminishing p62 & LC3 protein levels. Additionally, DAPA significantly lowered MDA content, increased GSH level and SOD activity. Moreover, it augmented the cytoprotective Nrf2/HO-1 signaling pathway. Furthermore, it attenuated renal and testicular apoptotic cell death via decreasing caspase-3 expression. Conclusion: Boosting autophagic events and combating oxidative stress and apoptosis by DAPA were engaged in alleviating Cd-induced renal and testicular impairment. This was accomplished by modulating the AMPK/mTOR and enhancing the Nrf2/HO-1 pathways.
Collapse
Affiliation(s)
- Doha M Dagher
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Marwa S Zaghloul
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura National University, Gamasa, Egypt
| | - Ghada M Suddek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
93
|
Zeng L, Zhu L, Fu S, Li Y, Hu K. Mitochondrial Dysfunction-Molecular Mechanisms and Potential Treatment approaches of Hepatocellular Carcinoma. Mol Cell Biochem 2025; 480:2131-2142. [PMID: 39463200 DOI: 10.1007/s11010-024-05144-4] [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: 08/06/2024] [Accepted: 10/18/2024] [Indexed: 10/29/2024]
Abstract
Primary liver cancer (PLC), also known as hepatocellular carcinoma (HCC), is a common type of malignant tumor of the digestive system. Its pathological form has a significant negative impact on the patients' quality of life and ability to work, as well as a significant financial burden on society. Current researches had identified chronic hepatitis B virus infection, aflatoxin B1 exposure, and metabolic dysfunction-associated steatotic liver disease (MASLD) as the main causative factors of HCC. Numerous variables, including inflammatory ones, oxidative stress, apoptosis, autophagy, and others, have been linked to the pathophysiology of HCC. On the other hand, autoimmune regulation, inflammatory response, senescence of the hepatocytes, and mitochondrial dysfunction are all closely related to the pathogenesis of HCC. In fact, a growing number of studies have suggested that mitochondrial dysfunction in hepatocytes may be a key factor in the pathogenesis of HCC. In disorders linked to cancer, mitochondrial dysfunction has gained attention in recent 10 years. As the primary producer of adenosine triphosphate (ATP) in liver cells, mitochondria are essential for preserving cell viability and physiological processes. By influencing multiple pathological processes, including mitochondrial fission/fusion, mitophagy, cellular senescence, and cell death, mitochondrial dysfunction contributes to the development of HCC. We review the molecular mechanisms of HCC-associated mitochondrial dysfunction and discuss new directions for quality control of mitochondrial disorders as a treatment for HCC.
Collapse
Affiliation(s)
- Lianlin Zeng
- Department of Rehabilitation Medicine, Suining Central Hospital, Suining, Sichuan Provience, China
| | - Lutao Zhu
- Department of Rehabilitation Medicine, Suining Central Hospital, Suining, Sichuan Provience, China
| | - Shasha Fu
- Department of Rehabilitation Medicine, Suining Central Hospital, Suining, Sichuan Provience, China
| | - Yangan Li
- Department of Rehabilitation Medicine, Suining Central Hospital, Suining, Sichuan Provience, China
| | - Kehui Hu
- Department of Rehabilitation Medicine, Suining Central Hospital, Suining, Sichuan Provience, China.
| |
Collapse
|
94
|
Yang B, Shen M, Lu C, Wang Y, Zhao X, Zhang Q, Qin X, Pei J, Wang H, Wang J. RNF144A inhibits autophagy by targeting BECN1 for degradation during L. monocytogenes infection. Autophagy 2025; 21:789-806. [PMID: 39608349 PMCID: PMC11925115 DOI: 10.1080/15548627.2024.2429380] [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: 04/19/2024] [Revised: 11/05/2024] [Accepted: 11/11/2024] [Indexed: 11/30/2024] Open
Abstract
Listeria monocytogenes (L. monocytogenes, Lm) is widely used in the laboratory as an infection model for the research on pathogenesis and host defense against gram-positive intracellular bacteria. Macroautophagy (called simply "autophagy" hereafter), is important in the host defense against pathogens, such as bacteria, viruses, and parasites. BECN1 plays a pivotal role in the initiation of autophagy and accumulating evidence indicates that post-translational modifications of BECN1 provide multiple strategies for autophagy regulation. In this study, we demonstrated that the RING1-IBR-RING2 (RBR) family member RNF144A (ring finger protein 144A), which was induced by Lm infection, promoted Lm infection in an autophagy-dependent but STING1-independent pattern. rnf144a deficiency in mice protected mice from Lm infection with inhibited innate immune responses. Interestingly, RNF144A decreased Lm-induced autophagosome accumulation. Mechanistic investigation indicated that RNF144A interacted with BECN1 and promoted its K48-linked ubiquitination, leading to the subsequent proteasome-dependent degradation of BECN1 and reduced autophagosome accumulation. Further study demonstrated that RNF144A promoted the ubiquitination of BECN1 at K117 and K427, and these two ubiquitination sites were essential to the role of BECN1 in autophagy and Lm infection. Thus, our findings suggested a new regulator in intracellular bacterial infection and autophagy, which may contribute to our understanding of host defense against intracellular bacterial infection via autophagy.Abbreviations: ATG3: autophagy related 3; ATG5: autophagy related 5; ATG7: autophagy related 7; ATG10: autophagy related 10; ATG12: autophagy related 12; ATG16L1: autophagy related 16 like 1; Baf A1: bafilomycin A1; BECN1: beclin 1; BMDC: bone marrow-derived dendritic cell; BMDM: bone marrow-derived macrophage; CFUs: colony-forming units; CHX: cycloheximide; CQ: chloroquine; CXCL10/IP-10: C-X-C motif chemokine ligand 10; EBSS: Earle's balanced salt solution; ELISA: enzyme-linked immunosorbent assay; IFIT1/ISG56: interferon induced protein with tetratricopeptide repeats 1; IFNB/IFN-β: interferon beta; IL6: interleukin 6; IRF3, interferon regulatory factor 3; Lm: L. monocytogenes; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MEF: mouse embryonic fibroblast; MOI: multiplicity of infection; PLA: proximity ligation assay; PMA: phorbol myristate acetate; PMA-THP1, PMA-differentiated THP1; PMs: peritoneal macrophages; PTMs: posttranslational modifications; RBR: RING1-IBR-RING2; RNF144A: ring finger protein 144A; STING1, stimulator of interferon response cGAMP interactor 1; TBK1, TANK binding kinase 1; TNF/TNF-α: tumor necrosis factor.
Collapse
Affiliation(s)
- Bo Yang
- Xinxiang Key Laboratory of Inflammation and Immunology, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Mengyang Shen
- Xinxiang Key Laboratory of Inflammation and Immunology, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Chen Lu
- Xinxiang Key Laboratory of Inflammation and Immunology, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yi Wang
- Xinxiang Key Laboratory of Inflammation and Immunology, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Xin Zhao
- Clinical Laboratory, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Qunmei Zhang
- Clinical Laboratory, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, China
| | - Xiao Qin
- Xinxiang Key Laboratory of Inflammation and Immunology, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Jinyong Pei
- Xinxiang Key Laboratory of Inflammation and Immunology, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Hui Wang
- Henan Key Laboratory of Immunology and Targeted Drug, Xinxiang Medical University, Xinxiang, Henan, China
| | - Jie Wang
- Xinxiang Key Laboratory of Inflammation and Immunology, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
| |
Collapse
|
95
|
Peng Y, She X, Peng Y. Characterization of key genes and immune cell infiltration associated with endometriosis through integrating bioinformatics and experimental analyses. Hereditas 2025; 162:49. [PMID: 40165344 PMCID: PMC11956255 DOI: 10.1186/s41065-025-00417-4] [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: 12/24/2024] [Accepted: 03/15/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUNDS Endometriosis (EM) is the most common gynecological disease in women of childbearing age. This study aims to identify key genes and screen drugs that may contribute to EM treatment. METHODS The differentially expressed genes (DEGs) were identified using limma analysis in the GSE11691 dataset. The protein-protein network (PPI) was constructed. Four machine learning methods, including LASSO, SVM-RFE, random forest, and Boruta, were applied to identify the key genes associated with EM. Flow cytometry, wound healing, and migration assays were applied to assess the cell functions of APLNR on hEM15A. The immune cell infiltration of each sample in EM was calculated using a single-sample gene set enrichment analysis (ssGSEA) algorithm. The potential drugs were screened using the Connectivity Map (CMAP) database, based on the DEGs. Finally, the expression levels of the three genes were further validated in the GSE23339 dataset. RESULTS One hundred thirty-seven down-regulated genes and 304 up-regulated genes were identified. We identified three key genes associated with EM: APLNR, HLA-DPA1, and AP1S2. The ssGSEA analysis results indicated that these genes play an important role in the development of EM. Moreover, EM immune cell infiltration was tightly associated with these three genes. Finally, several molecular compounds targeting EM were screened with the connectivity map (CMAP) database. ShAPLNR decreased the cell viability of hEM15A, increased the number of apoptotic cells, and significantly decreased the proportion of callus through APLNR in vitro studies. DISCUSSION Three genes (APLNR, HLA-DPA1, and AP1S2) may serve as novel therapeutic targets for diagnosing and treating patients with EM.
Collapse
Affiliation(s)
- Ying Peng
- Department of Obstetrics and Gynecology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
| | - Xiangdong She
- Department of Obstetrics and Gynecology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
| | - Ying Peng
- Department of Obstetrics and Gynecology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
96
|
Boggio V, Gonzalez CD, Zotta E, Ropolo A, Vaccaro MI. VMP1 Constitutive Expression in Mice Dampens Pancreatic and Systemic Histopathological Damage in an Experimental Model of Severe Acute Pancreatitis. Int J Mol Sci 2025; 26:3196. [PMID: 40243995 PMCID: PMC11988950 DOI: 10.3390/ijms26073196] [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: 02/17/2025] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
Acute pancreatitis (AP) an inflammatory condition caused by the premature activation of pancreatic proteases, leads to organ damage, systemic inflammation, and multi-organ failure. Severe acute pancreatitis (SAP) has high morbidity and mortality, affecting the liver, kidneys, and lungs. Autophagy maintains pancreatic homeostasis, with VMP1-mediated selective autophagy (zymophagy) preventing intracellular zymogen activation and acinar cell death. This study examines the protective role of VMP1 (Vacuole Membrane Protein 1)-induced autophagy using ElaI-VMP1 transgenic mice in a necrohemorrhagic SAP model (Hartwig's model). ElaI-VMP1 mice show significantly reduced pancreatic injury, including lower necrosis, edema, and inflammation, compared to wild-type (WT) mice. Biochemical markers (lactate dehydrogenase-LDH-, amylase, and lipase) and histopathology confirm that VMP1 expression mitigates pancreatic damage. Increased zymophagy negatively correlates with acinar necrosis, reinforcing its protective role. Beyond the pancreas, ElaI-VMP1 mice exhibit preserved liver, kidney, and lung histology, indicating reduced systemic organ damage. The liver maintains normal architecture, kidneys show minimal tubular necrosis, and lung inflammation features are reduced compared to WT mice. Our results confirm that zymophagy functions as a protective pathophysiological mechanism against pancreatic and extrapancreatic tissue injury in SAP. Further studies on the mechanism of VMP1-mediated selective autophagy in AP are necessary to determine its relevance and possible modulation to prevent the severity of AP.
Collapse
Affiliation(s)
- Veronica Boggio
- Instituto de Bioquímica y Biología Molecular Prof. Alberto Boveris, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires, Buenos Aires 1113, Argentina; (V.B.); (E.Z.)
| | - Claudio Daniel Gonzalez
- Centro de Educación Medica e Investigaciones Clínicas (CEMIC), Hospital Universitario Saavedra, Buenos Aires 1431, Argentina;
| | - Elsa Zotta
- Instituto de Bioquímica y Biología Molecular Prof. Alberto Boveris, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires, Buenos Aires 1113, Argentina; (V.B.); (E.Z.)
| | - Alejandro Ropolo
- Instituto de Bioquímica y Biología Molecular Prof. Alberto Boveris, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires, Buenos Aires 1113, Argentina; (V.B.); (E.Z.)
| | - Maria Ines Vaccaro
- Instituto de Bioquímica y Biología Molecular Prof. Alberto Boveris, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires, Buenos Aires 1113, Argentina; (V.B.); (E.Z.)
- Centro de Educación Medica e Investigaciones Clínicas (CEMIC), Hospital Universitario Saavedra, Buenos Aires 1431, Argentina;
| |
Collapse
|
97
|
Gambarotto L, Wosnitzka E, Nikoletopoulou V. The Life and Times of Brain Autophagic Vesicles. J Mol Biol 2025:169105. [PMID: 40154918 DOI: 10.1016/j.jmb.2025.169105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/17/2025] [Accepted: 03/22/2025] [Indexed: 04/01/2025]
Abstract
Most of the knowledge on the mechanisms and functions of autophagy originates from studies in yeast and other cellular models. How this valuable information is translated to the brain, one of the most complex and evolving organs, has been intensely investigated. Fueled by the tight dependence of the mammalian brain on autophagy, and the strong links of human brain diseases with autophagy impairment, the field has revealed adaptations of the autophagic machinery to the physiology of neurons and glia, the highly specialized cell types of the brain. Here, we first provide a detailed account of the tools available for studying brain autophagy; we then focus on the recent advancements in understanding how autophagy is regulated in brain cells, and how it contributes to their homeostasis and integrated functions. Finally, we discuss novel insights and open questions that the new knowledge has raised in the field.
Collapse
Affiliation(s)
- Lisa Gambarotto
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Erin Wosnitzka
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | | |
Collapse
|
98
|
Gatica D, Alsaadi RM, El Hamra R, Li B, Mueller R, Miyazaki M, Sun Q, Sad S, Russell RC. The ER-phagy receptor FAM134B is targeted by Salmonella Typhimurium to promote infection. Nat Commun 2025; 16:2923. [PMID: 40133256 PMCID: PMC11937434 DOI: 10.1038/s41467-025-58035-7] [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: 04/02/2024] [Accepted: 03/06/2025] [Indexed: 03/27/2025] Open
Abstract
Macroautophagy/autophagy is a key catabolic-recycling pathway that can selectively target damaged organelles or invading pathogens for degradation. The selective autophagic degradation of the endoplasmic reticulum (hereafter referred to as ER-phagy) is a homeostatic mechanism, controlling ER size, the removal of misfolded protein aggregates, and organelle damage. ER-phagy can also be stimulated by pathogen infection. However, the link between ER-phagy and bacterial infection remains poorly understood, as are the mechanisms evolved by pathogens to escape the effects of ER-phagy. Here, we show that Salmonella enterica serovar Typhimurium inhibits ER-phagy by targeting the ER-phagy receptor FAM134B, leading to a pronounced increase in Salmonella burden after invasion. Salmonella prevents FAM134B oligomerization, which is required for efficient ER-phagy. FAM134B knock-out raises intracellular Salmonella number, while FAM134B activation reduces Salmonella burden. Additionally, we found that Salmonella targets FAM134B through the bacterial effector SopF to enhance intracellular survival through ER-phagy inhibition. Furthermore, FAM134B knock-out mice infected with Salmonella presented severe intestinal damage and increased bacterial burden. These results provide mechanistic insight into the interplay between ER-phagy and bacterial infection, highlighting a key role for FAM134B in innate immunity.
Collapse
Affiliation(s)
- Damián Gatica
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Reham M Alsaadi
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Rayan El Hamra
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Boran Li
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Rudolf Mueller
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Makoto Miyazaki
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado Denver, Aurora, CO, USA
| | - Qiming Sun
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang, China
- Department of Biochemistry and Department of Cardiology, Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Subash Sad
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Ryan C Russell
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Canada.
- University of Ottawa Centre for Infection, Immunity and Inflammation, Ottawa, ON, Canada.
| |
Collapse
|
99
|
Coculo L, Wits M, Mariani I, Fianco G, Cappato S, Bocciardi R, Pedemonte N, Volpe E, Ciolfi S, Sessa RL, Rinaldo S, Cutruzzolà F, Trisciuoglio D, Goumans MJ, Sanchez-Duffhues G, Stagni V. Interplay between ALK2 R206H mutant receptor and autophagy signaling regulates receptor stability and its chondrogenic functions. Cell Death Discov 2025; 11:117. [PMID: 40121219 PMCID: PMC11929866 DOI: 10.1038/s41420-025-02393-0] [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: 08/08/2024] [Revised: 02/14/2025] [Accepted: 03/10/2025] [Indexed: 03/25/2025] Open
Abstract
Heterozygous mutations in the Bone morphogenetic protein (BMP) type I receptor ACVR1, encoding activin-like kinase 2 (ALK2), underlie all cases of the rare genetic musculoskeletal disorder Fibrodysplasia Ossificans Progressiva (FOP). The most commonly found mutant ALK2 p.R206H receptor variant exhibits loss of auto inhibition of BMP signaling and can be activated by Activins, while wild-type receptors remain unresponsive. Consequently, the downstream chondrogenic signaling is enhanced, thus driving heterotopic ossification within soft connective tissues. Despite several investigational treatments being evaluated in clinical trials, no cure for FOP exists today. The cellular and molecular mechanisms underlying disease progression are still being deciphered. In this study, we show a close interplay between the mutant ALK2R206H receptor signaling and dysregulation of the autophagic flux triggered by hypoxia. Mechanistically, reduced autophagic flux correlates with increased stability of ALK2R206H, resulting in sustained signaling. Of note, we demonstrated that Rapamycin, under clinical investigation as a treatment for FOP, inhibits chondrogenic differentiation in an autophagy-dependent manner. Consistently, other pharmacological autophagy inducers, like Spermidine, can reduce ALK2R206H driven chondrogenic differentiation in vitro. These results were verified in FOP patient-derived cells. In conclusion, this study shows that aberrant autophagic flux mediates sustained ALK2R206H signaling, introducing a novel druggable target in FOP by reactivating autophagy.
Collapse
Grants
- Seed Grant FOP GSA21A002 Fondazione Telethon (Telethon Foundation)
- Seed Grant FOP-Renewal GSA23I001 Fondazione Telethon (Telethon Foundation)
- Seed Grant FOP GSA21A002 Fondazione Telethon (Telethon Foundation)
- Seed Grant FOP-Renewal GSA23I001 Fondazione Telethon (Telethon Foundation)
- Seed Grant FOP-Renewal GSA23I001 Fondazione Telethon (Telethon Foundation)
- Seed Grant FOP GSA21A002 Fondazione Telethon (Telethon Foundation)
- PRIN202224M22R Ministero dell'Istruzione, dell'Università e della Ricerca (Ministry of Education, University and Research)
- PRIN202224M22R Ministero dell'Istruzione, dell'Università e della Ricerca (Ministry of Education, University and Research)
- Netherlands Cardiovascular Research Initiative (the Dutch Heart Foundation, Dutch Federation of University Medical Centers, the Netherlands Organization for Health Research and Development, and the Royal Netherlands Academy of Sciences), PHAEDRA-IMPACT (DCVA) and DOLPHIN-GENESIS (CVON).
- Ramón y Cajal RYC2021-030866-I, PID2022-141212OA-I00 and CNS2023-145432 from the Spanish Ministry of Science and Innovation GSD is also sponsored by La Marató de TV3 (202038-30), the BHF-DZHK-DHF, 2022/23 award PROMETHEUS, the Foundation Eugenio Rodriguez Pascual (FERP-2023-058) and the Foundation “Por dos pulgares de nada”
Collapse
Affiliation(s)
- Laura Coculo
- Institute of Molecular Biology and Pathology, National Research Council (CNR), Rome, Italy
- Cell Signalling Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Marius Wits
- Department of Cell & Chemical Biology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Irene Mariani
- Institute of Molecular Biology and Pathology, National Research Council (CNR), Rome, Italy
- Cell Signalling Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Giulia Fianco
- Institute of Molecular Biology and Pathology, National Research Council (CNR), Rome, Italy
| | - Serena Cappato
- UOC Genetica Medica, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Renata Bocciardi
- UOC Genetica Medica, IRCCS Istituto Giannina Gaslini, Genoa, Italy
- DINOGMI, University of Genoa, Genoa, Italy
| | | | - Elisabetta Volpe
- Molecular Neuroimmunology Unit, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Serena Ciolfi
- Molecular Neuroimmunology Unit, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Rosario Luigi Sessa
- Institute of Molecular Biology and Pathology, National Research Council (CNR), Rome, Italy
- Cell Signalling Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Serena Rinaldo
- Department of Biochemical Sciences, Sapienza University of Rome, Rome, Italy
| | | | - Daniela Trisciuoglio
- Institute of Molecular Biology and Pathology, National Research Council (CNR), Rome, Italy
| | - Marie-Josè Goumans
- Department of Cell & Chemical Biology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Gonzalo Sanchez-Duffhues
- Department of Cell & Chemical Biology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
- Nanomaterials and Nanotechnology Research Center (CINN), Spanish National Research Council (CSIC), Health Research Institute of Asturias (ISPA), Oviedo, Asturias, Spain
| | - Venturina Stagni
- Institute of Molecular Biology and Pathology, National Research Council (CNR), Rome, Italy.
- Cell Signalling Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy.
| |
Collapse
|
100
|
Zhou F, Deng S, Luo Y, Liu Z, Liu C. Research Progress on the Protective Effect of Green Tea Polyphenol (-)-Epigallocatechin-3-Gallate (EGCG) on the Liver. Nutrients 2025; 17:1101. [PMID: 40218859 PMCID: PMC11990830 DOI: 10.3390/nu17071101] [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: 02/24/2025] [Revised: 03/15/2025] [Accepted: 03/18/2025] [Indexed: 04/14/2025] Open
Abstract
The liver, as the primary metabolic organ, is susceptible to an array of factors that can harm liver cells and give rise to different liver diseases. Epigallocatechin gallate (EGCG), a natural compound found in green tea, exerts numerous beneficial effects on the human body. Notably, EGCG displays antioxidative, antibacterial, antiviral, anti-inflammatory, and anti-tumor properties. This review specifically highlights the pivotal role of EGCG in liver-related diseases, focusing on viral hepatitis, autoimmune hepatitis, fatty liver disease, and hepatocellular carcinoma. EGCG not only inhibits the entry and replication of hepatitis B and C viruses within hepatocytes, but also mitigates hepatocytic damage caused by hepatitis-induced inflammation. Furthermore, EGCG exhibits significant therapeutic potential against hepatocellular carcinoma. Combinatorial use of EGCG and anti-hepatocellular carcinoma drugs enhances the sensitivity of drug-resistant cancer cells to chemotherapeutic agents, leading to improved therapeutic outcomes. Thus, the combination of EGCG and anti-hepatocellular carcinoma drugs holds promise as an effective approach for treating drug-resistant hepatocellular carcinoma. In conclusion, EGCG possesses hepatoprotective properties against various forms of liver damage and emerges as a potential drug candidate for liver diseases.
Collapse
Affiliation(s)
- Fang Zhou
- School of Chemistry and Environmental Sciences, Xiangnan University, Chenzhou 423000, China;
| | - Sengwen Deng
- School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan 411201, China; (S.D.); (C.L.)
| | - Yong Luo
- School of Chemistry and Environmental Sciences, Xiangnan University, Chenzhou 423000, China;
| | - Zhonghua Liu
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China;
| | - Changwei Liu
- School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan 411201, China; (S.D.); (C.L.)
| |
Collapse
|