1
|
Zhang Z, Shen Z, Xie S, Li J, Zhang Z, Zhang S, Peng B, Huang Q, Li M, Ma S, Huang Q. Rapamycin exerts neuroprotective effects by inhibiting FKBP12 instead of mTORC1 in the mouse model of Parkinson's disease. Neuropharmacology 2025; 275:110504. [PMID: 40345576 DOI: 10.1016/j.neuropharm.2025.110504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 04/28/2025] [Accepted: 05/05/2025] [Indexed: 05/11/2025]
Abstract
Parkinson's disease (PD), characterized by the selective loss of nigral dopaminergic neurons, is a common neurodegenerative disorder for which effective disease-modifying therapies remain unavailable. Rapamycin, a clinical immunosuppressant used for decades, has demonstrated neuroprotective effects in various animal models of neurological diseases, including PD. These effects are believed to be mediated through the inhibition of mammalian target of rapamycin (mTOR) complex 1 (mTORC1) signaling, with rapamycin binding to FKBP12. However, recent studies have suggested that mTOR activation can be neuroprotective in degenerating dopaminergic neurons, presenting a paradox to the neuroprotective mechanism of rapamycin via mTORC1 inhibition. In this study, we showed that mTORC1 signaling was inactivated in nigral dopaminergic neurons in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD. Notably, the optimal neuroprotective dose of rapamycin did not inhibit mTORC1 signaling nor restore autophagy defects in nigral dopaminergic neurons of MPTP-treated male C57BL/6 mice. Furthermore, acute Raptor knockout in dopaminergic neurons, which abolishes mTORC1 activity, did not diminish rapamycin's neuroprotective effects, suggesting that its protection is independent of mTORC1 inhibition. Importantly, rapamycin is also a potent inhibitor of FKBP12, a peptidyl-prolyl cis-trans isomerase highly expressed in the brain. Selective knockdown of FKBP12 in nigral dopaminergic neurons confers neuroprotective effects comparable to that of rapamycin, with no synergism observed when the two are combined. Collectively, our results indicate that rapamycin exerts neuroprotective effects in parkinsonian mice through inhibition of FKBP12 rather than mTORC1 signaling. These findings suggest that FKBP12 may serve as a novel target for disease-modifying therapies in PD.
Collapse
Affiliation(s)
- Zeyan Zhang
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Ziyue Shen
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Shiming Xie
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Junyu Li
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Zeyu Zhang
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Sheng Zhang
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Bo Peng
- Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai, 200032, China
| | - Qianchu Huang
- Department of Psychology, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Mingtao Li
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Shanshan Ma
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan 2nd Road, Guangzhou, 510080, China.
| | - Qiaoying Huang
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, No. 74 Zhongshan 2nd Road, Guangzhou, 510080, China.
| |
Collapse
|
2
|
Kawahara T, Inazu T, Mizuno S, Tominaga N, Toda M, Toyama N, Kawahara C, Suzuki G. Anti-sarcopenic effects of active vitamin D through modulation of anabolic and catabolic signaling pathways in human skeletal muscle: A randomized controlled trial. Metabolism 2025; 168:156240. [PMID: 40158795 DOI: 10.1016/j.metabol.2025.156240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 03/14/2025] [Accepted: 03/22/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND The muscle-building and strengthening effects of the active form of vitamin D in humans remain unclear. METHODS In this ancillary study of the Diabetes Prevention with active Vitamin D trial, we examined clinical and experimental aspects to investigate the effects and mechanisms of eldecalcitol, an active form of vitamin D, in preventing sarcopenia. We examined changes in molecules involved in muscle synthesis and degradation pathways in muscle samples from 32 participants before and after 1 year of eldecalcitol or placebo treatment. The protein levels of molecules involved in muscle synthesis and degradation pathways were examined using western blotting. Additionally, the skeletal muscle and body fat volumes were measured using bioelectrical impedance analysis with a body composition analyzer. RESULTS We found that eldecalcitol treatment for 1 year resulted in higher phosphorylation levels of mTOR and FOXO1 signaling pathways, which are associated with increased muscle mass and strength than those with placebo treatment. Body composition measurements at 1 year showed that the eldecalcitol group had significantly higher skeletal muscle mass (1.9 % vs. -3.4 %, p = 3.26E-9) and muscle strength (4.1 % vs. -0.7 %, p = 2.57E-17), and lower fat mass (-3.2 % vs. 1.8 %, p = 1.73E-12) than those in the placebo group. CONCLUSION This study suggested that the active form of vitamin D regulates the protein synthesis and degradation pathways in human skeletal muscle and may help prevent sarcopenia. This study was registered at UMIN clinical trials registry, UMIN 000005394.
Collapse
Affiliation(s)
- Tetsuya Kawahara
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka 807-8555, Japan; Division of Endocrinology and Metabolism, Shinkomonji Hospital, Kitakyushu, Fukuoka 800-0057, Japan.
| | - Tetsuya Inazu
- Department of Pharmacy, College of Pharmaceutical Science, Ritsumeikan University, Kusatsu, Shiga 525-8577, Japan
| | - Shoichi Mizuno
- Division of Cancer Immunotherapy, National Cancer Center EPOC, Kashiwa, Chiba 277-8577, Japan
| | - Naoki Tominaga
- Division of Endocrinology and Metabolism, Shinkomonji Hospital, Kitakyushu, Fukuoka 800-0057, Japan
| | - Mikio Toda
- Division of Endocrinology and Metabolism, Shinkomonji Hospital, Kitakyushu, Fukuoka 800-0057, Japan
| | - Nagahiro Toyama
- Division of Endocrinology and Metabolism, Shinkomonji Hospital, Kitakyushu, Fukuoka 800-0057, Japan
| | - Chie Kawahara
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka 807-8555, Japan
| | - Gen Suzuki
- Department of Internal Medicine, International University Health and Welfare Clinic, Ohtawara, Tochigi 324-8501, Japan
| |
Collapse
|
3
|
O'Leary KM, Slezak T, Kossiakoff AA. Conformation-specific synthetic intrabodies modulate mTOR signaling with subcellular spatial resolution. Proc Natl Acad Sci U S A 2025; 122:e2424679122. [PMID: 40489625 DOI: 10.1073/pnas.2424679122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 04/02/2025] [Indexed: 06/11/2025] Open
Abstract
Subcellular compartmentalization is integral to the spatial regulation of mechanistic target of rapamycin (mTOR) signaling. However, the biological outputs associated with location-specific mTOR signaling events are poorly understood and challenging to decouple. Here, we engineered synthetic intracellular antibodies (intrabodies) that are capable of modulating mTOR signaling with genetically programmable spatial resolution. Epitope-directed phage display was exploited to generate high affinity synthetic antibody fragments (Fabs) against the FKBP12-Rapamycin binding site of mTOR (mTORFRB). We determined high-resolution crystal structures of two unique Fabs that discriminate distinct conformational states of mTORFRB through recognition of its substrate recruitment interface. By leveraging these conformation-specific binders as intracellular probes, we uncovered the structural basis for an allosteric mechanism governing mTOR complex 1 (mTORC1) stability mediated by subtle structural adjustments within mTORFRB. Furthermore, our results demonstrated that synthetic binders emulate natural substrates by employing divergent yet complementary hydrophobic residues at defined positions, underscoring the broad molecular recognition capability of mTORFRB. Intracellular signaling studies showed differential time-dependent inhibition of S6 kinase 1 and Akt phosphorylation by genetically encoded intrabodies, thus supporting a mechanism of inhibition analogous to the natural product rapamycin. Finally, we implemented a feasible approach to selectively modulate mTOR signaling in the nucleus through spatially programmed intrabody expression. These findings establish intrabodies as versatile tools for dissecting the conformational regulation of mTORC1 and should be useful to explore how location-specific mTOR signaling influences disease progression.
Collapse
Affiliation(s)
- Kelly M O'Leary
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60637
| | - Tomasz Slezak
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60637
| | - Anthony A Kossiakoff
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60637
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637
| |
Collapse
|
4
|
Kwakye J, Ariyo OW, Ghareeb AFA, Hartono E, Aryal B, Sovi S, Milfort MC, Fuller AL, Rekaya R, Aggrey SE. Effect of glucose supplementation on protein biosynthesis in chickens reared under thermoneutral or heat stress environment. Gene 2025; 951:149408. [PMID: 40064307 DOI: 10.1016/j.gene.2025.149408] [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: 10/01/2024] [Revised: 01/13/2025] [Accepted: 03/06/2025] [Indexed: 03/14/2025]
Abstract
Heat stress (HS) impacts broilers by reducing feed intake which impairs nutrient availability and energy levels, subsequently affecting protein biosynthesis. We hypothesize that an exogenous supply of glucose could provide extra energy resources that enhance protein biosynthesis in broilers reared under HS. Our experimental design involved two levels of temperature (25 °C [thermoneutral, TN]); 35 °C (8.00 AM to 8.00 PM, [Heat Stress, HS]), and two glucose levels (0 % and 6 %). We randomly assigned a total of 456 four-week-old Cobb500 broilers to four different treatment groups (TN0, TN6, HS0, and HS6), respectively. After 7 days post-HS, we observed an inverse relationship between the avian target of rapamycin (avTOR) and autophagy-related genes. The phosphorylation of mTOR and S6K1 at Ser2448 and Thr421/Ser424 respectively was higher (p < 0.05) in the TN0 group than in the HS groups. Additionally, the phosphorylation of Foxo3a at Ser253 was higher (p < 0.05) in the HS0 group than in the HS6 groups, indicating an adaptive response to HS. Thus, the combined effect of HS and glucose could influence the phosphorylation status of key signaling genes in the mTOR pathway. The expression levels of mRNA genes in the mTOR pathway were more pronounced (p < 0.05) in HS6 birds than in HS0 birds except for avTOR, Akt1, and S6K1.
Collapse
Affiliation(s)
- Josephine Kwakye
- NutriGenomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| | - Oluwatomide W Ariyo
- NutriGenomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| | - Ahmed F A Ghareeb
- NutriGenomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| | - Evan Hartono
- NutriGenomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| | - Bikash Aryal
- NutriGenomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| | - Selorm Sovi
- NutriGenomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| | - Marie C Milfort
- NutriGenomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| | - Alberta L Fuller
- NutriGenomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| | - Romdhane Rekaya
- Department of Animal and Dairy Science, University of Georgia, Athens, GA 30602, USA
| | - Samuel E Aggrey
- NutriGenomics Laboratory, Department of Poultry Science, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
5
|
Evans JF, Ledwell OA, Tang Y, Rue R, Mukhitov AR, Diesler R, Lin SM, Vanka KS, Basil MC, Cantu E, Henske EP, Krymskaya VP. The Bi-steric Inhibitor RMC-5552 Reduces mTORC1 Signaling and Growth in Lymphangioleiomyomatosis. Am J Respir Cell Mol Biol 2025; 72:643-652. [PMID: 39531634 DOI: 10.1165/rcmb.2024-0242oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 11/12/2024] [Indexed: 11/16/2024] Open
Abstract
Mutations in the TSC (tuberous sclerosis complex) genes result in the hyperactivation of the mTORC1 (mechanistic/mammalian target of rapamycin 1) growth pathway in mesenchymal pulmonary cells. Rapamycin (sirolimus), a naturally occurring macrolide, is the only therapeutic approved for women with lymphangioleiomyomatosis (LAM), a progressive, destructive lung disease caused by TSC gene mutations and mTORC1 hyperactivation. However, on cessation of the drug, lung function decline continues. We demonstrated here that pulmonary LAM cancer stem-like state (SLS) cells most highly expressed the eIF4E (eukaryotic translation initiation factor 4E)-dependent translation initiation genes. We also showed that the 4E-BP1 (eukaryotic initiation factor 4E-binding protein 1) gene has the lowest expression in these cells, indicating that the 4E-BP1/eIF4E ratio in LAM SLS cells favors unrestrained eIF4E oncogenic mRNA translation. The bi-steric mTORC1-selective compound RMC-5552 prevented growth of LAM-associated fibroblasts and phosphorylation of proteins in the ribosomal protein S6K1/ribosomal protein S6 (S6K1/S6) and 4E-BP1/eIF4E translation mTORC1-driven pathways, whereas rapamycin only blocked the S6K/S6 axis. Rapamycin inhibition of LAM-associated fibroblast growth was rapidly reversed, but RMC-5552 inhibition was more durable. RMC-5552, through its potential to eradicate LAM cancer SLS cells, may have therapeutic benefit in LAM and other diseases with mTORC1 hyperactivity.
Collapse
Affiliation(s)
- Jilly F Evans
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
- Lung Biology Institute, Perelman School of Medicine, and
| | - Owen A Ledwell
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
- Lung Biology Institute, Perelman School of Medicine, and
| | - Yan Tang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Ryan Rue
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
- Lung Biology Institute, Perelman School of Medicine, and
| | - Alexander R Mukhitov
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
- Lung Biology Institute, Perelman School of Medicine, and
| | - Rémi Diesler
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; and
- Department of Respiratory Medicine, National Reference Centre for Rare Pulmonary Diseases, Hospices Civils de Lyon, Université Lyon 1, UMR754, INRAE, ERN-LUNG, Lyon, France
| | - Susan M Lin
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
- Lung Biology Institute, Perelman School of Medicine, and
| | - Kanth Swaroop Vanka
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
- Lung Biology Institute, Perelman School of Medicine, and
| | - Maria C Basil
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
- Lung Biology Institute, Perelman School of Medicine, and
| | - Edward Cantu
- Division of Cardiovascular Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Elizabeth P Henske
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Vera P Krymskaya
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
- Lung Biology Institute, Perelman School of Medicine, and
| |
Collapse
|
6
|
Gao R, Shi J, Lyu Y, Ren B, Wei W, Cheng J, Chen J, Zhou Y, Chen J, Sun X, Jiang J, Li B, Yang K. ALKBH5 Regulates Macrophage Senescence and Accelerates Atherosclerosis by Promoting CCL5 m 6A Modification. Arterioscler Thromb Vasc Biol 2025; 45:928-944. [PMID: 40177773 DOI: 10.1161/atvbaha.125.322508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Accepted: 03/20/2025] [Indexed: 04/05/2025]
Abstract
BACKGROUND Senescent foamy macrophages are key drivers of atherosclerosis and plaque instability. N6-methyladenosine (m6A) modification of RNA plays an important role in the development of various diseases including aging. Here, we aim to investigate the role of m6A modification of RNA in the formation of senescent foamy macrophages in atherosclerosis. METHODS To assess m6A methylation, macrophages were isolated from the atherosclerotic plaques of patients with atherosclerosis, and Apoe-/- mice were fed a high-fat diet using flow cytometry. An ALKBH5 (alkB homolog 5)f/f, Lyz2 (lysozyme 2)Cre, Apoe-/- mouse model was generated to determine the infiltration of senescent foamy macrophages into plaques and atherosclerosis progression. Methylated RNA immunoprecipitation, RNA immunoprecipitation sequencing, and dual-luciferase assays were performed to explore the mechanisms underlying the ALKBH5-mediated formation of senescent foamy macrophages. RESULTS Decreased m6A methylation and increased ALKBH5 expression were observed in arterial plaques and infiltrating macrophages from patients and mice with atherosclerosis. Compared with control mice, ALKBH5f/f, Lyz2Cre, Apoe-/- mice exhibited fewer atherosclerosis plaques with greater stability, which was attributed to the suppression of senescent foamy macrophage formation and senescence-associated secretory phenotype. In addition, ALKBH5 deletion reduced the mRNA expression level of CCL5 (CC chemokine ligand 5) by increasing m6A methylation in macrophages, which disrupts the stability of CCL5 mRNA. Mechanistically, ALKBH5 promoted senescent foamy macrophage formation through the CCL5/CCR5 (CC chemokine receptor 5)/autophagy signaling pathway. CCL5 also recruited CD8+ IFN (interferon)γ+ T cells via the CCL5-CCR5 axis. The ALKBH5 inhibitor IOX1 (inhibitor of 2OG oxygenases) and the CCR5 antagonist maraviroc were identified as potential clinical interventions for inhibiting senescent foamy macrophage formation and atherosclerosis progression. CONCLUSIONS Myeloid ALKBH5 deletion attenuates atherosclerosis progression by suppressing the formation of senescent foamy macrophages and the recruitment of CD8+IFNγ+ T cells. These findings identify ALKBH5, CCL5, and CCR5 as novel therapeutic targets for atherosclerosis.
Collapse
Affiliation(s)
- Rifeng Gao
- Department of Cardiac Surgery (R.G., Jianxin Chen, J.J., K.Y.), The Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Jiaran Shi
- Department of Cardiology, Lihuili Hospital Facilitated to Ningbo University, China (J.S.)
| | - Yang Lyu
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University, China (Y.L.)
| | - Bichen Ren
- Department of Vascular Surgery, Institute of Vascular Surgery (B.R.), Zhongshan Hospital, Fudan University, China
| | - Wei Wei
- Department of Cardiology, Shanghai Chest Hospital (W.W.), Shanghai Jiao Tong University, China
| | - Jiahui Cheng
- Department of Radiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, China (J.C., Y.Z., B.L.)
| | - Juntao Chen
- Department of Urology (Juntao Chen), The Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Yan Zhou
- Department of Radiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, China (J.C., Y.Z., B.L.)
| | - Jianxin Chen
- Department of Cardiac Surgery (R.G., Jianxin Chen, J.J., K.Y.), The Second Affiliated Hospital, Zhejiang University, Hangzhou, China
- Department of Cardiovascular Medicine, Guangxin District Traditional Chinese Medicine Hospital, Jiangxi, China (Jianxin Chen)
| | - Xiaolei Sun
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases (X.S.), Zhongshan Hospital, Fudan University, China
| | - Jun Jiang
- Department of Cardiac Surgery (R.G., Jianxin Chen, J.J., K.Y.), The Second Affiliated Hospital, Zhejiang University, Hangzhou, China
- Department of Cardiology (J.J.), The Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Bo Li
- Department of Radiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, China (J.C., Y.Z., B.L.)
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, China (B.L.)
| | - Kun Yang
- Department of Cardiac Surgery (R.G., Jianxin Chen, J.J., K.Y.), The Second Affiliated Hospital, Zhejiang University, Hangzhou, China
- Department of Cardiology, Ruijin Hospital, School of Medicine (K.Y.), Shanghai Jiao Tong University, China
| |
Collapse
|
7
|
Rao G, Sang X, Zhu X, Zou S, Zhang Y, Cheng W, Tian Y, Fu X. Pathological Glucose Levels Enhance Entry Factor Expression and Hepatic SARS-CoV-2 Infection. J Cell Mol Med 2025; 29:e70581. [PMID: 40442985 PMCID: PMC12122388 DOI: 10.1111/jcmm.70581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 04/17/2025] [Accepted: 04/23/2025] [Indexed: 06/02/2025] Open
Abstract
Accumulating clinical evidence suggests an intricate relationship between severe COVID-19 and preexisting metabolic complications, which share some metabolic dysregulations, including hyperglycaemia, hyperinsulinaemia and hyperlipidaemia. However, the potential role of these metabolic risk factors in SARS-CoV-2 infection and entry factor expression remains unknown. Here we report the implication of hyperglycaemia in SARS-CoV-2 infection and therapy. Hyperglycaemia, instead of hyperinsulinaemia and hyperlipidaemia, can significantly induce the expression of SARS-CoV-2 entry factors (Ace2, Tmprss2, Tmprss4, Furin and Nrp1) in liver cells, but not in lung and pancreatic cells, which is attenuated by mTOR inhibition. Correspondingly, pathological glucose levels promote SARS-CoV-2 entry into cultured hepatocytes in pseudovirus cell systems. Conversely, representative glucose-lowering drugs (metformin, dapagliflozin, sitagliptin and exenatide) are able to diminish the enhancement of entry factor expression and SARS-CoV-2 infection in cultured hepatocytes under pathological glucose conditions. Intriguingly, SARS-CoV-2 entry factors are increased in the livers of nonalcoholic fatty liver disease and diabetes patients. These results define hyperglycaemia as a key susceptibility factor for hepatic SARS-CoV-2 infection, and provide insights into the clinical application of glucose-lowering therapies in COVID-19 patients under comorbid hyperglycaemia conditions.
Collapse
Affiliation(s)
- Guocheng Rao
- Department of Endocrinology and Metabolism, Department of Biotherapy, Center for Diabetes and Metabolism Research, State Key Laboratory of Biotherapy and Cancer Center, West China HospitalSichuan UniversityChengduSichuanChina
| | - Xiongbo Sang
- Department of Endocrinology and Metabolism, Department of Biotherapy, Center for Diabetes and Metabolism Research, State Key Laboratory of Biotherapy and Cancer Center, West China HospitalSichuan UniversityChengduSichuanChina
| | - Xinyue Zhu
- Department of Endocrinology and Metabolism, Department of Biotherapy, Center for Diabetes and Metabolism Research, State Key Laboratory of Biotherapy and Cancer Center, West China HospitalSichuan UniversityChengduSichuanChina
| | - Sailan Zou
- Department of Endocrinology and Metabolism, Department of Biotherapy, Center for Diabetes and Metabolism Research, State Key Laboratory of Biotherapy and Cancer Center, West China HospitalSichuan UniversityChengduSichuanChina
| | - Yanyan Zhang
- Department of Endocrinology and Metabolism, Department of Biotherapy, Center for Diabetes and Metabolism Research, State Key Laboratory of Biotherapy and Cancer Center, West China HospitalSichuan UniversityChengduSichuanChina
- Department of Endocrinology and MetabolismGansu Provincial HospitalLanzhouChina
| | - Wei Cheng
- Division of Pulmonary and Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China HospitalSichuan UniversityChengduSichuanChina
| | - Yan Tian
- Department of Endocrinology and Metabolism, Department of Biotherapy, Center for Diabetes and Metabolism Research, State Key Laboratory of Biotherapy and Cancer Center, West China HospitalSichuan UniversityChengduSichuanChina
| | - Xianghui Fu
- Department of Endocrinology and Metabolism, Department of Biotherapy, Center for Diabetes and Metabolism Research, State Key Laboratory of Biotherapy and Cancer Center, West China HospitalSichuan UniversityChengduSichuanChina
| |
Collapse
|
8
|
He D, Dai W, Cui Y, Gao R, Yang X, Li W, Zhou J, Yin S, Kuang T, Zhu J, Luo L, Wang R, Qu Y, Yun C, Liu Z, Diao X, Ma X, Liang H, Wang F. Diamine oxidase acts as a novel risk factor in abnormal inflammation via mediating "cytosolic ROS-autophagy-IFN-γ" axis in NK cells. Life Sci 2025; 377:123775. [PMID: 40449879 DOI: 10.1016/j.lfs.2025.123775] [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: 01/12/2025] [Revised: 05/21/2025] [Accepted: 05/27/2025] [Indexed: 06/03/2025]
Abstract
AIMS Diamine oxidase (DAO), a well-established biomarker for intestinal damage, histamine intolerance or tumorigenesis, has rarely been reported in immune regulation. This study aimed to identify DAO as a critical enhancer of abnormal inflammation by promoting interferon-gamma (IFN-γ) production from natural killer (NK) cells. MAIN METHODS Clinical bioinformatics analyzed aoc1 (DAO-coding gene) expression in PBMCs from patients with inflammatory diseases. Murine models (LPS-induced systemic inflammation, sepsis, DSS-induced colitis) using DAO-/- mice, alongside DAO-/- NK92 cells and DAO inhibitor DIZE, were employed for phenotypic validation. Cellular profiling, bone marrow chimeras, reciprocal transplantation, RNA-sequence, non-targeted metabolomics, and flow cytometry were utilized to dissect DAO's mechanisms in NK cells. KEY FINDINGS DAO deficiency protected mice from inflammatory pathology by suppressing IFN-γ production. NK cells were identified as the primary target cells during the process, with DAO acting intracellularly to promote IFN-γ via a reactive oxygen species (ROS)-autophagy axis. DAO-derived ROS, distinct from mitochondrial or NOX2 sources, enhanced autophagic flux during NK activation, enabling IFN-γ biosynthesis. DAO did not affect NK homeostasis, including maturation, proliferation, or receptor expressions. SIGNIFICANCE DAO is a novel risk factor in inflammatory diseases, driving IFN-γ production through ROS-autophagy signaling in NK cells. Targeting DAO may offer therapeutic strategies for conditions involving dysregulated IFN-γ responses, including sepsis, colitis, and autoimmune disorders.
Collapse
Affiliation(s)
- Dongmei He
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China
| | - Weihong Dai
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China; Emergency of The Second Affiliated Hospital, Hainan Medical University, Haikou 571100, Hainan, China
| | - Yiqin Cui
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China
| | - Rui Gao
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China
| | - Xue Yang
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China
| | - Wei Li
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China
| | - Jing Zhou
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China
| | - Shuangqin Yin
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China
| | - Tianyin Kuang
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China
| | - Junyu Zhu
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China
| | - Li Luo
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China
| | - Rixing Wang
- Emergency of The Second Affiliated Hospital, Hainan Medical University, Haikou 571100, Hainan, China
| | - Ye Qu
- Emergency of The Second Affiliated Hospital, Hainan Medical University, Haikou 571100, Hainan, China
| | - Caihong Yun
- Emergency of The Second Affiliated Hospital, Hainan Medical University, Haikou 571100, Hainan, China
| | - Zhuli Liu
- Emergency of The Second Affiliated Hospital, Hainan Medical University, Haikou 571100, Hainan, China
| | - Xiaoyan Diao
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China
| | - Xiaoyuan Ma
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China.
| | - Huaping Liang
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China.
| | - Fangjie Wang
- The First Research Department, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center of PLA, Chongqing 400042, China.
| |
Collapse
|
9
|
Mantoan Ritter L, Annear NMP, Baple EL, Ben-Chaabane LY, Bodi I, Brosson L, Cadwgan JE, Coslett B, Crosby AH, Davies DM, Daykin N, Dedeurwaerdere S, Dühring Fenger C, Dunlop EA, Elmslie FV, Girodengo M, Hambleton S, Jansen AC, Johnson SR, Kearley KC, Kingswood JC, Laaniste L, Lachlan K, Latchford A, Madsen RR, Mansour S, Mihaylov SR, Muhammed L, Oliver C, Pepper T, Rawlins LE, Schim van der Loeff I, Siddiqui A, Takhar P, Tatton-Brown K, Tee AR, Tibarewal P, Tye C, Ultanir SK, Vanhaesebroeck B, Zare B, Pal DK, Bateman JM. mTOR pathway diseases: challenges and opportunities from bench to bedside and the mTOR node. Orphanet J Rare Dis 2025; 20:256. [PMID: 40426219 PMCID: PMC12107773 DOI: 10.1186/s13023-025-03740-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 04/16/2025] [Indexed: 05/29/2025] Open
Abstract
Mechanistic target of rapamycin (mTOR) is a highly conserved serine/threonine kinase that regulates key cellular processes including cell growth, autophagy and metabolism. Hyperactivation of the mTOR pathway causes a group of rare and ultrarare genetic diseases. mTOR pathway diseases have diverse clinical manifestations that are managed by distinct medical disciplines but share a common underlying molecular basis. There is a now a deep understanding of the molecular underpinning that regulates the mTOR pathway but effective treatments for most mTOR pathway diseases are lacking. Translating scientific knowledge into clinical applications to benefit the unmet clinical needs of patients is a major challenge common to many rare diseases. In this article we expound how mTOR pathway diseases provide an opportunity to coordinate basic and translational disease research across the group, together with industry, medical research foundations, charities and patient groups, by pooling expertise and driving progress to benefit patients. We outline the germline and somatic mutations in the mTOR pathway that cause rare diseases and summarise the prevalence, genetic basis, clinical manifestations, pathophysiology and current treatments for each disease in this group. We describe the challenges and opportunities for progress in elucidating the underlying mechanisms, improving diagnosis and prognosis, as well as the development and approval of new therapies for mTOR pathway diseases. We illustrate the crucial role of patient public involvement and engagement in rare disease and mTOR pathway disease research. Finally, we explain how the mTOR Pathway Diseases node, part of the Research Disease Research UK Platform, will address these challenges to improve the understanding, diagnosis and treatment of mTOR pathway diseases.
Collapse
Affiliation(s)
- Laura Mantoan Ritter
- King's College London Institute of Psychiatry Psychology and Neuroscience, London, UK
- King's College Hospital NHS Foundation Trust, London, UK
| | - Nicholas M P Annear
- St George's University Hospitals NHS Foundation Trust, London, UK
- School of Health & Medical Sciences, City St George's, University of London, London, UK
| | | | - Leila Y Ben-Chaabane
- King's College London Institute of Psychiatry Psychology and Neuroscience, London, UK
| | - Istvan Bodi
- King's College Hospital NHS Foundation Trust, London, UK
| | | | | | | | | | | | | | | | | | | | - Frances V Elmslie
- St George's University Hospitals NHS Foundation Trust, London, UK
- School of Health & Medical Sciences, City St George's, University of London, London, UK
| | - Marie Girodengo
- King's College London Institute of Psychiatry Psychology and Neuroscience, London, UK
- The Francis Crick Institute, London, UK
| | - Sophie Hambleton
- Newcastle University Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | | | - Simon R Johnson
- Centre for Respiratory Research, NIHR Nottingham Biomedical Research Centre and Biodiscovery Institute, Translational Medical Sciences, University of Nottingham, Nottingham, UK
| | - Kelly C Kearley
- mTOR Node Advisory Panel (MAP), London, UK
- PTEN UK and Ireland Patient Group, London, UK
| | - John C Kingswood
- St George's University Hospitals NHS Foundation Trust, London, UK
| | | | - Katherine Lachlan
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Andrew Latchford
- Polyposis Registry, St Mark's Hospital, London, UK
- Department of Surgery and Cancer, Imperial College London, London, UK
| | | | - Sahar Mansour
- St George's University Hospitals NHS Foundation Trust, London, UK
- School of Health & Medical Sciences, City St George's, University of London, London, UK
| | | | | | | | - Tom Pepper
- PTEN Research, Cheltenham, Gloucestershire, UK
| | | | - Ina Schim van der Loeff
- Newcastle University Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Ata Siddiqui
- King's College Hospital NHS Foundation Trust, London, UK
| | | | - Katrina Tatton-Brown
- St George's University Hospitals NHS Foundation Trust, London, UK
- School of Health & Medical Sciences, City St George's, University of London, London, UK
| | | | | | - Charlotte Tye
- King's College London Institute of Psychiatry Psychology and Neuroscience, London, UK
| | | | | | | | - Deb K Pal
- King's College London Institute of Psychiatry Psychology and Neuroscience, London, UK
| | - Joseph M Bateman
- King's College London Institute of Psychiatry Psychology and Neuroscience, London, UK.
| |
Collapse
|
10
|
Kraft M, Schoofs H, Petkova M, Andrade J, Grosso AR, Benedito R, De Roo AK, Boon LM, Vikkula M, Kapp FG, Hägerling R, Potente M, Mäkinen T. Angiopoietin-TIE2 feedforward circuit promotes PIK3CA-driven venous malformations. NATURE CARDIOVASCULAR RESEARCH 2025:10.1038/s44161-025-00655-9. [PMID: 40410415 DOI: 10.1038/s44161-025-00655-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 04/11/2025] [Indexed: 05/25/2025]
Abstract
Venous malformations (VMs) are vascular anomalies lacking curative treatments, often caused by somatic PIK3CA mutations that hyperactivate the PI3Kα-AKT-mTOR signaling pathway. Here, we identify a venous-specific signaling circuit driving disease progression, where excessive PI3Kα activity amplifies upstream TIE2 receptor signaling through autocrine and paracrine mechanisms. In Pik3caH1047R-driven VM mouse models, single-cell transcriptomics and lineage tracking revealed clonal expansion of mutant endothelial cells with a post-capillary venous phenotype, characterized by suppression of the AKT-inhibited FOXO1 and its target genes, including the TIE2 antagonist ANGPT2. An imbalance in TIE2 ligands, likely exacerbated by aberrant recruitment of smooth muscle cells producing the agonist ANGPT1, increased TIE2 activity in both mouse and human VMs. While mTOR blockade had limited effects on advanced VMs in mice, inhibiting TIE2 or ANGPT effectively suppressed their growth. These findings uncover a PI3K-FOXO1-ANGPT-TIE2 circuit as a core driver of PIK3CA-related VMs and highlight TIE2 as a promising therapeutic target.
Collapse
Affiliation(s)
- Marle Kraft
- Uppsala University, Department of Immunology, Genetics and Pathology, Uppsala, Sweden
| | - Hans Schoofs
- Uppsala University, Department of Immunology, Genetics and Pathology, Uppsala, Sweden
| | - Milena Petkova
- Uppsala University, Department of Immunology, Genetics and Pathology, Uppsala, Sweden
| | - Jorge Andrade
- Angiogenesis & Metabolism Laboratory, Center of Vascular Biomedicine, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Ana Rita Grosso
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Lisbon, Portugal
- UCIBIO - Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Rui Benedito
- Molecular Genetics of Angiogenesis Group. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - An-Katrien De Roo
- Center for Vascular Anomalies, VASCERN VASCA European Reference Center, Cliniques Universitaires Saint Luc, UCLouvain, Brussels, Belgium
- Department of Pathology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
- Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
| | - Laurence M Boon
- Center for Vascular Anomalies, VASCERN VASCA European Reference Center, Cliniques Universitaires Saint Luc, UCLouvain, Brussels, Belgium
- Division of Plastic Surgery, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
- Laboratory of Human Molecular Genetics, de Duve Institute, UCLouvain, Brussels, Belgium
| | - Miikka Vikkula
- Center for Vascular Anomalies, VASCERN VASCA European Reference Center, Cliniques Universitaires Saint Luc, UCLouvain, Brussels, Belgium
- Laboratory of Human Molecular Genetics, de Duve Institute, UCLouvain, Brussels, Belgium
- WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Friedrich G Kapp
- Department of Pediatric Hematology and Oncology, Children's Hospital, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, VASCERN VASCA European Reference Center, Freiburg, Germany
| | - René Hägerling
- Research Group 'Lymphovascular Medicine and Translational 3D-Histopathology', Institute of Medical and Human Genetics, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Center for Regenerative Therapies, Berlin, Germany
| | - Michael Potente
- Angiogenesis & Metabolism Laboratory, Center of Vascular Biomedicine, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Taija Mäkinen
- Uppsala University, Department of Immunology, Genetics and Pathology, Uppsala, Sweden.
- Translational Cancer Medicine Program and Department of Biochemistry and Developmental Biology, University of Helsinki, Helsinki, Finland.
- Wihuri Research Institute, Helsinki, Finland.
| |
Collapse
|
11
|
Zhang F, Xu J, Yuan Q, Xie Y, Zhang L, Zhang J, Xu Z, Liu M. Deletion of ZNRF2 Exacerbates MPTP-Induced Parkinson's Disease by Activating mTOR-Mediated Neuroinflammatory Pathways. Mol Neurobiol 2025:10.1007/s12035-025-05044-8. [PMID: 40402410 DOI: 10.1007/s12035-025-05044-8] [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: 11/21/2024] [Accepted: 05/06/2025] [Indexed: 05/23/2025]
Abstract
Parkinson's disease (PD) imposes a significant health burden among older adults and may be related to zinc and ring finger 2 (ZNRF2)-a member of the ubiquitination family. To investigate the role and mechanism of action of ZNRF2 in the regulation of mammalian target of rapamycin (mTOR)-mediated neuroinflammation in a mouse model of PD. Healthy mice were injected intraperitoneally with either saline (control) or MPTP 30 mg/kg. Mouse behavior was tested using rotarod and open field tests. The distribution and expression of tyrosine hydroxylase (TH) were determined by immunoblotting and immunohistochemistry. Inflammatory factors were evaluated using immunoblotting, enzyme-linked immunosorbent assay, and immunofluorescence assay. Compared with mice injected with saline, MPTP-treated mice showed significantly impaired locomotor activity, a significant decrease in the number of TH neurons, and a markedly altered morphology. ZNRF2 expression was significantly increased in the mesencephalon of MPTP-treated mice compared to that in control mice. ZNRF2 knockdown exacerbated motor dysfunction, accelerated dopamine neuron degeneration and death, increased the levels of pro-inflammatory factors (e.g., interleukin (IL)-1β, IL-6), and suppressed the expression of anti-inflammatory factors (e.g., IL-4, IL-10) in the central nervous system of MPTP-treated mice, with more pronounced activation of microglia and astrocytes. ZNRF2 knockdown significantly elevated phosphorylated mTOR protein levels after MPTP treatment; subsequently, phosphorylated mTOR protein levels were inhibited; dyskinesia and dopamine neuronal damage were significantly ameliorated, and neuroinflammation was suppressed in PD mice. ZNRF2 regulates the pathogenesis of MPTP-induced PD in mice via mechanisms related to mTOR-mediated neuroinflammation.
Collapse
Affiliation(s)
- Fanshi Zhang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Huichuan District, No. 149, Dalian Road, Zunyi, 563000, China
| | - Jingqing Xu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Huichuan District, No. 149, Dalian Road, Zunyi, 563000, China
| | - Qi Yuan
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Huichuan District, No. 149, Dalian Road, Zunyi, 563000, China
| | - Yuling Xie
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Huichuan District, No. 149, Dalian Road, Zunyi, 563000, China
| | - Li Zhang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Huichuan District, No. 149, Dalian Road, Zunyi, 563000, China
| | - Jun Zhang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Huichuan District, No. 149, Dalian Road, Zunyi, 563000, China.
| | - Zucai Xu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Huichuan District, No. 149, Dalian Road, Zunyi, 563000, China.
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi, China.
| | - Mei Liu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Huichuan District, No. 149, Dalian Road, Zunyi, 563000, China.
| |
Collapse
|
12
|
Ren S, Zhu J, Shan G, Liang J, Bian Y, Lin H, Shi H, Pan B, Zhao G, Yang H, Huang X, Zhan C, Ge D, Bi G. Transcription factor ZNF266 suppresses cancer progression by modulating CA9-mediated intracellular pH alteration in lung adenocarcinoma. Respir Res 2025; 26:191. [PMID: 40389968 PMCID: PMC12090625 DOI: 10.1186/s12931-025-03278-7] [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: 01/11/2025] [Accepted: 05/15/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND Lung cancer remains the leading cause of cancer-related mortality globally, with lung adenocarcinoma (LUAD) being the most prevalent subtype. Despite extensive research efforts, the role of transcription factors in LUAD progression remains largely uncharacterized. In this study, we focused on ZNF266, a transcription factor whose impacts on LUAD have not been investigated. METHODS Using high-throughput sequencing data, we observed a significant downregulation of ZNF266 expression in LUAD tissues. To validate this finding, we conducted a retrospective analysis of nearly three thousand LUAD patients' data from public databases and our institution. Functional studies were performed using cell lines, organoids, and xenograft models to assess the role of ZNF266 in LUAD progression. RNA sequencing, chromatin immunoprecipitation, DNA pull-down assays, and dual-luciferase reporter assays were employed to elucidate the underlying mechanism. Additionally, adeno-associated virus (AAV)-mediated overexpression of ZNF266 was used to evaluate its therapeutic potential. RESULTS Patients with low ZNF266 expression had poorer prognosis compared to those with high expression. ZNF266 inhibits the malignant phenotypes of LUAD, including proliferation, migration, and invasion. Mechanistically, ZNF266 binds to the promoter region of CA9, suppressing its transcription. This leads to a reduction in intracellular pH and subsequent inhibition of the mTOR signaling pathway, which is crucial for cancer cell growth and survival. Furthermore, AAV-mediated overexpression of ZNF266 significantly inhibited tumor growth in patient-derived xenograft models. CONCLUSIONS Our study demonstrated that ZNF266 inhibits LUAD progression in a pH-dependent manner via modulating CA9 expression, uncovering its therapeutic significance for LUAD treatment.
Collapse
Affiliation(s)
- Shencheng Ren
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Junkan Zhu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Guangyao Shan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jiaqi Liang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yunyi Bian
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Han Lin
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Haochun Shi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Binyang Pan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Guangyin Zhao
- Department of Thoracic Surgery, Shanghai Geriatric Medical Center, Fudan University, Shanghai, 201104, China
| | - Huiqin Yang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xiaolong Huang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Cheng Zhan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Di Ge
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Guoshu Bi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
13
|
Hou T, Yan P, Inuzuka H, Wei W. Lysosomal EGFR functions as a GEF for Rheb. Cell Res 2025:10.1038/s41422-025-01126-3. [PMID: 40335660 DOI: 10.1038/s41422-025-01126-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2025] Open
Affiliation(s)
- Tao Hou
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Peiqiang Yan
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Hiroyuki Inuzuka
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
14
|
Han F, Simeroth S, Zhu J, Gryniuk I, Pranay A, Chen W, Wang Y, Cai Y, Shen Z, Wang G, Griffin CT, Xia L, Yu P. Lymphatic endothelial mTORC1 instructs metabolic and developmental signaling during lymphangiogenesis. Dev Cell 2025:S1534-5807(25)00250-3. [PMID: 40339577 DOI: 10.1016/j.devcel.2025.04.012] [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: 12/07/2023] [Revised: 11/10/2024] [Accepted: 04/16/2025] [Indexed: 05/10/2025]
Abstract
The lymphatic vasculature comprises lymphatic capillaries and collecting vessels. To support lymphatic development, lymphatic endothelial cells (LECs) utilize nutrients to fuel lymphangiogenic processes. Meanwhile, LECs maintain constant prospero homeobox 1 (PROX1) expression critical for lymphatic specification. However, molecular mechanisms orchestrating nutrient metabolism while sustaining PROX1 levels in LECs remain unclear. Here, we show that loss of RAPTOR, an indispensable mechanistic target of rapamycin complex 1 (mTORC1) component, downregulates PROX1 and impairs lymphatic capillary growth and differentiation of collecting lymphatics in mice. Mechanistically, mTORC1 inhibition in mouse and human LECs causes Myc reduction, which decreases hexokinase 2 (HK2) and glutaminase (GLS), inhibiting glycolysis and glutaminolysis. Myc or HK2/GLS ablation impedes lymphatic capillary and collecting vessel formation. Interestingly, mTORC1 regulation of PROX1 is independent of Myc-HK2/GLS signaling. Moreover, genetic interaction analysis indicates that Myc and PROX1 play crucial roles in mTORC1-regulated lymphatic development. Collectively, our findings identify mTORC1 as a key regulator of metabolic programs and PROX1 expression during lymphangiogenesis.
Collapse
Affiliation(s)
- Fei Han
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Summer Simeroth
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Jie Zhu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Irma Gryniuk
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Atul Pranay
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Weiqing Chen
- Center for Bioinformatics and Computational Biology, Houston Methodist Research Institute, Houston, TX, USA; Department of Physiology, Biophysics & Systems Biology, Weill Cornell Graduate School of Medical Science, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Yuan Wang
- Department of Radiation Oncology, Rutgers Cancer Institute and Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Yuanyuan Cai
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Zhiyuan Shen
- Department of Radiation Oncology, Rutgers Cancer Institute and Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Guangyu Wang
- Center for Bioinformatics and Computational Biology, Houston Methodist Research Institute, Houston, TX, USA; Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, USA; Center for RNA Therapeutics, Houston Methodist Research Institute, Houston, TX, USA; Department of Cardiothoracic Surgery, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Courtney T Griffin
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Lijun Xia
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; Department of Biochemistry and Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Pengchun Yu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| |
Collapse
|
15
|
Niu P, Li D, Chen H, Zhu Y, Zhou J, Zhang J, Liu Y. Cardamonin suppresses mTORC1/SREBP1 through reducing Raptor and inhibits de novo lipogenesis in ovarian cancer. PLoS One 2025; 20:e0322733. [PMID: 40315213 PMCID: PMC12047825 DOI: 10.1371/journal.pone.0322733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 03/25/2025] [Indexed: 05/04/2025] Open
Abstract
Metabolic reprogramming is a hallmark of cancer and de novo lipogenesis (DNL) accelerates the progression of ovarian cancer. In this study, we investigated the effects of cardamonin, a natural compound potential to suppress various malignancies, on the lipid anabolism in ovarian cancer. Cell proliferation was assessed using CCK-8 and clone formation assay. Cell apoptosis was detected by flow cytometry with Annexin V-FITC/PI staining and mitochondrial membrane potential (MMP) was measured with JC-10 probe. Free fatty acids (FFA) was measured by fluorescence using acyl-CoA oxidation and carnitine palmitoyl transferase-1 (CPT-1) activity was analyzed by spectrophotometric assay using palmitoyl-CoA and DTNB (5,5'-dithio-bis-(2-nitrobenzoic acid)) reaction. mRNA expression was measured by Quantitative Real-Time PCR. Protein expression was analyzed through western blotting and immunofluorescence. Raptor was knocked down by shRNA and Raptor was overexpressed by lentiviral transfection. The antitumor effect of cardamonin was evaluated using a xenotransplantation tumor bearing mouse model. Cardamonin suppressed the cell proliferation, induced cell apoptosis and triggered mitochondrial damage in ovarian cancer cells. Cardamonin inhibited the protein expression of sterol regulatory element binding protein 1 (SREBP1) and its downstream lipogenic enzymes and decreased FFA content and CPT-1 activity. Additionally, cardamonin inhibited the activation of mechanistic target of rapamycin complex 1 (mTORC1) and expression of regulatory-associated protein of mTOR (Raptor). Raptor knockdown abolished the inhibitory effect of cardamonin on mTORC1 and SREBP1. Furthermore, cardamonin inhibited mTORC1 activation and lipogenic proteins expression induced by Raptor overexpression. Cardamonin reduced the tumor growth and fatty acid synthase of the tumors, as evidenced by decreased expression of Ki-67 and FASN. It suggests that cardamonin suppresses mTORC1/SREBP1 through reducing the protein level of Raptor and inhibits DNL of ovarian cancer.
Collapse
Affiliation(s)
- Peiguang Niu
- Department of Pharmacy, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Key Laboratory of Women and Children’s Critical Diseases Research [Fujian Maternity and Child Health Hospital (Fujian Women and Children’s Hospital)], Fujian Maternity and Child Health Hospital, Fuzhou, Fujian, China
| | - Danyun Li
- Department of Pharmacy, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China
| | - Huajiao Chen
- Department of Pharmacy, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China
| | - Yanting Zhu
- Department of Pharmacy, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Key Laboratory of Women and Children’s Critical Diseases Research [Fujian Maternity and Child Health Hospital (Fujian Women and Children’s Hospital)], Fujian Maternity and Child Health Hospital, Fuzhou, Fujian, China
| | - Jintuo Zhou
- Department of Pharmacy, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China
| | - Jinhua Zhang
- Department of Pharmacy, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China
| | - Ying Liu
- Department of Pharmacy, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
16
|
McKenzie CI, Dvorscek AR, Ding Z, Robinson MJ, O'Donnell K, Pitt C, Ferguson DT, Mulder J, Herold MJ, Tarlinton DM, Quast I. Syndecans and glycosaminoglycans influence B-cell development and activation. EMBO Rep 2025; 26:2435-2458. [PMID: 40155751 PMCID: PMC12069707 DOI: 10.1038/s44319-025-00432-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 03/07/2025] [Accepted: 03/12/2025] [Indexed: 04/01/2025] Open
Abstract
Syndecans (SDCs) are glycosaminoglycan-containing cell surface proteins with diverse functions in the immune system with SDC1 (CD138) and SDC4 expressed in B-lineage cells. Here, we show that stem cells lacking either molecule generate fewer B-cell progenitors but give rise to mature B cells in vivo. Deletion of the plasma cell "marker" CD138 has no effect on homeostatic or antigen-induced plasma cell formation. Naive B cells express high SDC4 and encounter with cognate antigen results in transient CD138 upregulation and SDC4 loss, both further modulated by IL-4, IL-21, and CD40 ligation. SDC4 is downregulated on germinal center B cells and absent on most memory B cells. Glycosaminoglycans such as those attached to SDCs, and heparin, a commonly used therapeutic, regulate survival and activation of naive B cells by limiting responsiveness to cognate antigen. Conversely, ablation of SDC4 results in increased baseline and antigen-induced B-cell activation. Collectively, our data reveal B-cell activation- and subset-dependent SDC expression and show that SDC4 and GAGs can limit antigen-induced activation to promote B-cell survival and expansion.
Collapse
Affiliation(s)
- Craig I McKenzie
- Department of Immunology, Monash University, Melbourne, VIC, 3004, Australia.
- Murdoch Children's Research Institute, Melbourne, VIC, 3052, Australia.
| | - Alexandra R Dvorscek
- Department of Immunology, Monash University, Melbourne, VIC, 3004, Australia
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Zhoujie Ding
- Department of Immunology, Monash University, Melbourne, VIC, 3004, Australia
| | - Marcus J Robinson
- Department of Immunology, Monash University, Melbourne, VIC, 3004, Australia
| | - Kristy O'Donnell
- Department of Immunology, Monash University, Melbourne, VIC, 3004, Australia
| | - Catherine Pitt
- Department of Immunology, Monash University, Melbourne, VIC, 3004, Australia
| | - Daniel T Ferguson
- Australian Centre for Blood Diseases, Monash University, Melbourne, VIC, 3004, Australia
| | - Jesse Mulder
- Department of Immunology, Monash University, Melbourne, VIC, 3004, Australia
| | - Marco J Herold
- Blood Cells and Blood Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, 3052, Australia
- Olivia Newton-John Cancer Research Centre, Heidelberg, VIC, 3084, Australia
- School of Cancer Medicine, La Trobe University, Heidelberg, VIC, 3084, Australia
| | - David M Tarlinton
- Department of Immunology, Monash University, Melbourne, VIC, 3004, Australia
| | - Isaak Quast
- Department of Immunology, Monash University, Melbourne, VIC, 3004, Australia.
| |
Collapse
|
17
|
Hartung J, Müller C, Calkhoven CF. The dual role of the TSC complex in cancer. Trends Mol Med 2025; 31:452-465. [PMID: 39488444 DOI: 10.1016/j.molmed.2024.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/10/2024] [Accepted: 10/14/2024] [Indexed: 11/04/2024]
Abstract
The tuberous sclerosis complex (TSC1/TSC2/TBC1D7) primarily functions to inhibit the mechanistic target of rapamycin complex 1 (mTORC1), a crucial regulator of cell growth. Mutations in TSC1 or TSC2 cause tuberous sclerosis complex (TSC), a rare autosomal dominant genetic disorder marked by benign tumors in multiple organs that rarely progress to malignancy. Traditionally, TSC proteins are considered tumor suppressive due to their inhibition of mTORC1 and other mechanisms. However, more recent studies have shown that TSC proteins can also promote tumorigenesis in certain cancer types. In this review, we explore the composition and function of the TSC protein complex, the roles of its individual components in cancer biology, and potential future therapeutic targeting strategies.
Collapse
Affiliation(s)
- Josephine Hartung
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9700 AD Groningen, The Netherlands
| | - Christine Müller
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9700 AD Groningen, The Netherlands
| | - Cornelis F Calkhoven
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9700 AD Groningen, The Netherlands.
| |
Collapse
|
18
|
Bankolé A, Srivastava A, Shihavuddin A, Tighanimine K, Faucourt M, Koka V, Weill S, Nemazanyy I, Nelson AJ, Stokes MP, Delgehyr N, Genovesio A, Meunier A, Fumagalli S, Pende M, Spassky N. mTOR controls ependymal cell differentiation by targeting the alternative cell cycle and centrosomal proteins. EMBO Rep 2025:10.1038/s44319-025-00460-2. [PMID: 40307619 DOI: 10.1038/s44319-025-00460-2] [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: 10/21/2024] [Revised: 03/18/2025] [Accepted: 04/04/2025] [Indexed: 05/02/2025] Open
Abstract
Ependymal cells are multiciliated glial cells lining the ventricles of the mammalian brain. Their differentiation from progenitor cells involves cell enlargement and progresses through centriole amplification phases and ciliogenesis. These phases are accompanied by the sharp up-regulation of mTOR Complex 1 activity (mTORC1), a master regulator of macromolecule biosynthesis and cell growth, whose function in ependymal cell differentiation is unknown. We demonstrate that mTORC1 inhibition by rapamycin preserves the progenitor pool by reinforcing quiescence and preventing alternative cell cycle progression for centriole amplification. Overexpressing E2F4 and MCIDAS circumvents mTORC1-regulated processes, enabling centriole amplification despite rapamycin, and enhancing mTORC1 activity through positive feedback. Acute rapamycin treatment in multicentriolar cells during the late phases of differentiation causes centriole regrouping, indicating a direct role of mTORC1 in centriole dynamics. By phosphoproteomic and phosphomutant analysis, we reveal that the mTORC1-mediated phosphorylation of GAS2L1, a centrosomal protein that links actin and microtubule cytoskeletons, participates in centriole disengagement. This multilayered and sequential control of ependymal development by mTORC1, from the progenitor pool to centriolar function, has implications for pathophysiological conditions like aging and hydrocephalus-prone genetic diseases.
Collapse
Affiliation(s)
- Alexia Bankolé
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades-INEM, F-75015, Paris, France
| | - Ayush Srivastava
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Cilia Biology and neurogenesis, 75005, Paris, France
| | - Asm Shihavuddin
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Computational bioimaging and bioinformatics, 75005, Paris, France
- Department of EEE, Presidency University, Dhaka, Bangladesh
| | - Khaled Tighanimine
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades-INEM, F-75015, Paris, France
| | - Marion Faucourt
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Cilia Biology and neurogenesis, 75005, Paris, France
| | - Vonda Koka
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades-INEM, F-75015, Paris, France
| | - Solene Weill
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Cilia Biology and neurogenesis, 75005, Paris, France
| | - Ivan Nemazanyy
- Platform for Metabolic Analyses, Structure Fédérative de Recherche Necker, INSERM US24/CNRS UAR 3633, Paris, France
| | - Alissa J Nelson
- Cell Signaling Technology INC, 3 Trask Lane, Danvers, MA, 01923, USA
| | - Matthew P Stokes
- Cell Signaling Technology INC, 3 Trask Lane, Danvers, MA, 01923, USA
| | - Nathalie Delgehyr
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Cilia Biology and neurogenesis, 75005, Paris, France
| | - Auguste Genovesio
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Computational bioimaging and bioinformatics, 75005, Paris, France
| | - Alice Meunier
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Cilia Biology and neurogenesis, 75005, Paris, France
| | - Stefano Fumagalli
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades-INEM, F-75015, Paris, France
| | - Mario Pende
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades-INEM, F-75015, Paris, France.
| | - Nathalie Spassky
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Cilia Biology and neurogenesis, 75005, Paris, France.
| |
Collapse
|
19
|
Li Y, Lang M, He Q, Hu Y, Shi H, Zheng S, Wu Z, Zhou S. Nutritional and hormonal regulation of mitochondrial biogenesis drives fat body remodeling for reproductive competence. J Adv Res 2025:S2090-1232(25)00285-1. [PMID: 40306618 DOI: 10.1016/j.jare.2025.04.041] [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: 01/07/2025] [Revised: 04/04/2025] [Accepted: 04/26/2025] [Indexed: 05/02/2025] Open
Abstract
INTRODUCTION Insect fat body serves as a central hub for energy mobilization and protein synthesis. During larval metamorphosis, fat body undergoes programmed cell death and tissue disassembly. Following adult eclosion, fat body reconstructs with cell proliferation and becomes competent for large-scale vitellogenin (Vg) synthesis required for the maturation of dozens of eggs. OBJECTIVES This study aims to uncover the molecular mechanisms underlying the remodeling of fat body in acquisition of competence for massive Vg production. METHODS RNA-seq and metabolomics were used for identification of differentially expressed genes and metabolites. RNAi was applied for gene knockdown. Transmission electron microscope, MitoTracker staining, mitochondrial DNA quantification, ATP and citrate synthase assays were employed for examining mitochondrial biogenesis. Dual-luciferase reporter assay and EMSA were performed for transcriptional regulation. qRT-PCR and western blot were performed for measuring Vg synthesis. RESULTS Transcriptomic and metabolomic analyses revealed significant upregulation of genes and metabolites involved in mitochondrial biogenesis in the fat body of adult locusts. PGC-1α was highly expressed in adult fat body. Knockdown of PGC-1α reduced mitochondrial biogenesis, fat body cell number, Vg synthesis and ovarian development. CREBB bound to PGC-1α promoter and activated its transcription. CREBB depletion impaired mitochondrial biogenesis and fat body remodeling. Moreover, loss of TORC1 function suppressed CREBB function and PGC-1α expression, subsequently disrupting mitochondrial biogenesis and fat body remodeling. Juvenile hormone (JH) deprivation also decreased CREBB function and PGC-1α expression, which was reversible with JH treatment. Our results suggest that TORC1 and JH coordinate CREBB-upregulated PGC-1α expression, which promotes mitochondrial biogenesis and fat body remodeling for Vg synthesis and egg production. CONCLUSION The findings provide new insights into the molecular mechanisms of post-metamorphic fat body development, and highlight the role of JH/TORC1/CREBB/PGC-1α/mitochondrial biogenesis axis in insect reproduction. The data also offer potential targets for insect pest control.
Collapse
Affiliation(s)
- Yiying Li
- State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, School of Life Sciences, Henan University, Kaifeng, China
| | - Mengyao Lang
- State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, School of Life Sciences, Henan University, Kaifeng, China
| | - Qiongjie He
- State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, School of Life Sciences, Henan University, Kaifeng, China
| | - Yuanyuan Hu
- State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, School of Life Sciences, Henan University, Kaifeng, China
| | - Huanhuan Shi
- State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, School of Life Sciences, Henan University, Kaifeng, China
| | - Siqian Zheng
- State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, School of Life Sciences, Henan University, Kaifeng, China
| | - Zhongxia Wu
- State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, School of Life Sciences, Henan University, Kaifeng, China.
| | - Shutang Zhou
- State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, School of Life Sciences, Henan University, Kaifeng, China.
| |
Collapse
|
20
|
Chen X, Zhu Y, Xia L, Su S, Fan S, Lu Y, Chen Q, Wei Y, Huang Q, Liu X, Peng X. Glutamine limits NLRP3 inflammasome activation and pyroptosis in macrophages by sustaining the IRG1/itaconate axis. FEBS J 2025. [PMID: 40296302 DOI: 10.1111/febs.70119] [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: 09/02/2024] [Revised: 01/18/2025] [Accepted: 04/17/2025] [Indexed: 04/30/2025]
Abstract
Aberrant activation of NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) inflammasome increases the release of mature pro-inflammatory cytokines interleukin (IL)-1β and IL-18, and enhances pyroptosis; thereby necessitating tight regulation of the NLRP3 inflammasome. Dysfunctional glutamine metabolism contributes to the pathogenesis of multiple inflammatory disorders, and the precise mechanism remains to be elucidated. Here, we provide evidence that glutamine deprivation enhances NLRP3 inflammasome activation in macrophages. Indeed, the absence of exogenous glutamine specifically enhanced NLRP3 inflammasome assembly, thereby accelerating pyroptosis and promoting the maturation of IL-1β and IL-18. Inhibition of glutaminolysis exhibited a similar effect to glutamine deprivation, whereas this effect was reversed by α-ketoglutarate (α-KG), a tricarboxylic acid (TCA)-cycle intermediate that can be replenished by glutamine supply. We further observed reduced generation of endogenous itaconate by glutamine deprivation and verified that both exogenous supplementation of itaconate derivative and increased endogenous itaconate production by overexpressing immune-responsive gene 1 [IRG1; also known as aconitate decarboxylase 1 (ACOD1)] could replace glutamine to inhibit the NLRP3 inflammasome. Mechanistically, glutamine deprivation decreased the source of substrate and inhibited transcription factor EB (TFEB)-dependent transcriptional upregulation of IRG1, thereby impairing the IRG1/itaconate axis that suppresses the NLRP3 inflammasome. Furthermore, glutamine deficiency was detected in a murine sepsis model, whereas extrinsic glutamine supplementation conferred protection against intestinal inflammation and tissue damage in septic mice. Taken together, our findings provide a novel insight into the link between glutamine metabolism and NLRP3 inflammasome activation, highlighting the target of glutamine metabolism, which holds as a potential therapeutic strategy for inflammatory diseases.
Collapse
Affiliation(s)
- Xiaoli Chen
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yuanfeng Zhu
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Lin Xia
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Sen Su
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Shijun Fan
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yongling Lu
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Qian Chen
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yan Wei
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Qianying Huang
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xin Liu
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xi Peng
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
21
|
Li X, Han M, Zhu H, Pan Y, Su C, Liu Y, Liao Z, Zhang B, Chen X. m 6A-Mediated TMCO3 Promotes Hepatocellular Carcinoma Progression by Facilitating the Membrane Translocation and Activation of AKT. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2504187. [PMID: 40285646 DOI: 10.1002/advs.202504187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Indexed: 04/29/2025]
Abstract
The transmembrane and coiled-coil domains 3 (TMCO3) are highly expressed in many tumors. However, the underlying mechanisms governing the way in which TMCO3 affects the progression of hepatocellular carcinoma (HCC) remain unclear. This study screens out the molecule TMCO3 with high N6-methyladenosine (m6A) modification level in tumor samples compared to the adjacent non-cancerous tissues of three pairs of HCC patients through Methylated RNA Immunoprecipitation Sequencing (MeRIP-seq) and RNA sequencing (RNA-seq). Subsequently, the oncogenic effect of TMCO3 in HCC is verified through in vivo and in vitro experiments. AlkB Homolog 5 (ALKBH5), an m6A demethylase of TMCO3 is then screened out. The following experiments demonstrate that TMCO3 can activate AKT directly through the Phosphatidylinositol-3-Kinase (PI3K) pathway, thus promoting the progression of HCC. Meanwhile, the phosphorylation site on TMCO3: the 85th amino acid-serine, and mutation of this site can directly impair the activity and membrane translocation of AKT is found. Finally, the carcinogenic effect of TMCO3 is further elucidated in HCC through the orthotopic treatment model and the hydrodynamic tail vein injection treatment model. The findings can provide a potential target for targeted AKT treatment in patients with HCC and verify a possible prognostic marker in HCC.
Collapse
Affiliation(s)
- Xinxin Li
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, 430030, China
| | - Mengzhen Han
- Department of General Surgery, Ezhou Central Hospital, Ezhou, Hubei, 436099, China
| | - He Zhu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, 430030, China
| | - Yonglong Pan
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, 430030, China
| | - Chen Su
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, 430030, China
| | - Yachong Liu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, 430030, China
| | - Zhibin Liao
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, 430030, China
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, 430030, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, Hubei, 430030, China
| | - Xiaoping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, 430030, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, Hubei, 430030, China
| |
Collapse
|
22
|
Dong Z, She X, Ma J, Chen Q, Gao Y, Chen R, Qin H, Shen B, Gao H. The E3 Ligase NEDD4L Prevents Colorectal Cancer Liver Metastasis via Degradation of PRMT5 to Inhibit the AKT/mTOR Signaling Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2504704. [PMID: 40279519 DOI: 10.1002/advs.202504704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Indexed: 04/27/2025]
Abstract
Colorectal cancer is the second most common cause of cancer mortality worldwide, and liver metastasis is the major cause of death of patients with colorectal cancer. Dysfunctional E3 ligase activity has recently been shown to be associated with colorectal cancer. However, the key E3 ligases affecting colorectal cancer liver metastasis remain unknown. Therefore, an shRNA library targeting 156 E3 ubiquitin ligases has been used to perform an in vivo loss-of-function screen of a human colorectal cancer cell line in a mouse model of liver metastasis. The screen reveals that neural precursor cell expressed developmentally down-regulated gene 4-like (NEDD4L) knockdown promotes colorectal cancer liver metastasis. Mechanistic studies reveal that NEDD4L binds to the PPNAY motif in protein arginine methyltransferase 5 (PRMT5) and ubiquitinates PRMT5 to promote its degradation. PRMT5 degradation attenuates the arginine methylation of AKT1 to inhibit the AKT/mTOR signaling pathway. The effect of NEDD4L decreases colorectal cancer cell proliferation to suppress colonization. This study is the first to show that PRMT5 is a substrate of NEDD4L and reveals not only the metastasis-inhibiting function of NEDD4L but also a novel mechanism by which NEDD4L prevents colorectal cancer liver metastasis. These findings may provide a new preventive strategy for liver metastasis.
Collapse
Affiliation(s)
- Zhewen Dong
- Tongji University Cancer Center and Research Institute of Intestinal Diseases, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, P. R. China
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, P. R. China
| | - Xiaofei She
- Tongji University Cancer Center and Research Institute of Intestinal Diseases, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, P. R. China
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, P. R. China
| | - Junxian Ma
- Tongji University Cancer Center and Research Institute of Intestinal Diseases, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, P. R. China
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, P. R. China
| | - Qian Chen
- Tongji University Cancer Center and Research Institute of Intestinal Diseases, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, P. R. China
| | - Yaqun Gao
- Tongji University Cancer Center and Research Institute of Intestinal Diseases, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, P. R. China
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, P. R. China
| | - Ruiyan Chen
- Tongji University Cancer Center and Research Institute of Intestinal Diseases, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, P. R. China
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, P. R. China
| | - Huanlong Qin
- Tongji University Cancer Center and Research Institute of Intestinal Diseases, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, P. R. China
| | - Bing Shen
- Tongji University Cancer Center and Research Institute of Intestinal Diseases, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, P. R. China
- Department of Urology and Urologic Cancer Institute, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, P. R. China
| | - Hua Gao
- Tongji University Cancer Center and Research Institute of Intestinal Diseases, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, P. R. China
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, P. R. China
| |
Collapse
|
23
|
Park S, Jeong J, Ahn K. Human cytomegalovirus infection induces L1 expression through UL38-dependent mTOR-KAP1 pathway. PLoS One 2025; 20:e0320512. [PMID: 40267069 PMCID: PMC12017509 DOI: 10.1371/journal.pone.0320512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 02/19/2025] [Indexed: 04/25/2025] Open
Abstract
Human cytomegalovirus (HCMV) and LINE-1 (L1) can co-inhabit a common host and closely interact with each other within a single cell. We have previously shown that HCMV exploits this opportunistic interaction by upregulating L1 expression that promotes its own productive life cycle by facilitating HCMV DNA replication. However, the mechanism by which HCMV increases L1 expression remains unknown. Here, we report that HCMV infection functionally inactivates KRAB-associated protein 1 (KAP1), a key epigenetic repressor of L1, through phosphorylation. HCMV infection of cells activates mTOR kinase that phosphorylates S824 residue of KAP1 and reduces its epigenetic repressive function, leading to increased chromatin accessibility of L1 promoter region. Treatment of potent mTOR inhibitor to the HCMV-infected cells was sufficient to reduce KAP1 phosphorylation and block L1 expression. Furthermore, cells infected with a mutant virus lacking UL38, an HCMV mTOR pathway activator, showed reduced KAP1 S824 phosphorylation and abolished L1 expression. Our results highlight the synergistic interaction between HCMV and L1 where HCMV UL38 serves as a primary viral regulator of L1 expression by upregulating the mTOR-KAP1 pathway.
Collapse
Affiliation(s)
- Sehong Park
- Center for RNA Research, Institute for Basic Science, Seoul, Republic of Korea
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
- SNU Institute for Virus Research, Seoul National University, Seoul, Republic of Korea
| | - Jiseok Jeong
- Center for RNA Research, Institute for Basic Science, Seoul, Republic of Korea
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
- SNU Institute for Virus Research, Seoul National University, Seoul, Republic of Korea
| | - Kwangseog Ahn
- Center for RNA Research, Institute for Basic Science, Seoul, Republic of Korea
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
- SNU Institute for Virus Research, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
24
|
He X, Wang QX, Wei D, Lin Y, Zhang X, Wu Y, Qian X, Lin Z, Xiao B, Wu Q, Wang Z, Zhou F, Wei Z, Wang J, Gong R, Zhang R, Zhang Q, Ding K, Gao S, Kang T. Lysosomal EGFR acts as a Rheb-GEF independent of its kinase activity to activate mTORC1. Cell Res 2025:10.1038/s41422-025-01110-x. [PMID: 40259053 DOI: 10.1038/s41422-025-01110-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 03/19/2025] [Indexed: 04/23/2025] Open
Abstract
Oncogenic mutations in EGFR often result in EGF-independent constitutive activation and aberrant trafficking and are associated with several human malignancies, including non-small cell lung cancer. A major consequence of EGFR mutations is the activation of the mechanistic target of rapamycin complex 1 (mTORC1), which requires EGFR kinase activity and downstream PI3K/AKT signaling, resulting in increased cell proliferation. However, recent studies have elucidated kinase-independent roles of EGFR in cell survival and cancer progression. Here, we report a cis mTORC1 activation function of EGFR that is independent of its kinase activity. Our results reveal that lysosomal localization of EGFR is critical to mTORC1 activation, where EGFR physically binds Rheb, acting as a guanine exchange factor (GEF) for Rheb, with its Glu804 serving as a potential glutamic finger. Genetic knock-in of EGFR-E804K in cells reduces the level of GTP-bound Rheb, and significantly suppresses mTORC1 activation, cell proliferation and tumor growth. Different tyrosine kinase inhibitors exhibit distinct effects on EGFR-induced mTORC1 activation, with afatinib, which additionally blocks EGFR's GEF activity, causing a much greater suppression of mTORC1 activation and cell growth, and erlotinib, which targets only kinase activity, resulting in only a slight decrease. Moreover, a novel small molecule, BIEGi-1, was designed to target both the Rheb-GEF and kinase activities of EGFR, and shows a strong inhibitory effect on the viability of cells harboring EGFR mutants. These findings unveil a fundamental event in cell growth and suggest a promising strategy against cancers with EGFR mutations.
Collapse
Affiliation(s)
- Xiaobo He
- Sun Yat-sen University Cancer Center, Guangdong Provincial Clinical Research Center for Cancer, State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, China
| | - Qiu-Xia Wang
- Sun Yat-sen University Cancer Center, Guangdong Provincial Clinical Research Center for Cancer, State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, China
| | - Denghui Wei
- Sun Yat-sen University Cancer Center, Guangdong Provincial Clinical Research Center for Cancer, State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, China.
| | - Yujie Lin
- Sun Yat-sen University Cancer Center, Guangdong Provincial Clinical Research Center for Cancer, State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, China
| | - Xia Zhang
- Sun Yat-sen University Cancer Center, Guangdong Provincial Clinical Research Center for Cancer, State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, China
| | - Yuanzhong Wu
- Sun Yat-sen University Cancer Center, Guangdong Provincial Clinical Research Center for Cancer, State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, China
| | - Xuexia Qian
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Zhihao Lin
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Beibei Xiao
- Sun Yat-sen University Cancer Center, Guangdong Provincial Clinical Research Center for Cancer, State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, China
| | - Qinxue Wu
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Zhen Wang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Fengtao Zhou
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development, Ministry of Education (MoE) of People's Republic of China, College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - Zhihao Wei
- Sun Yat-sen University Cancer Center, Guangdong Provincial Clinical Research Center for Cancer, State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, China
- Department of Oncology Radiotherapy, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jingxuan Wang
- Sun Yat-sen University Cancer Center, Guangdong Provincial Clinical Research Center for Cancer, State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, China
| | - Run Gong
- Sun Yat-sen University Cancer Center, Guangdong Provincial Clinical Research Center for Cancer, State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, China
| | - Ruhua Zhang
- Sun Yat-sen University Cancer Center, Guangdong Provincial Clinical Research Center for Cancer, State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, China
| | - Qingling Zhang
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China.
| | - Ke Ding
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development, Ministry of Education (MoE) of People's Republic of China, College of Pharmacy, Jinan University, Guangzhou, Guangdong, China.
| | - Song Gao
- Sun Yat-sen University Cancer Center, Guangdong Provincial Clinical Research Center for Cancer, State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, China.
- Integrated Traditional Chinese and Western Medicine Research Center, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China.
| | - Tiebang Kang
- Sun Yat-sen University Cancer Center, Guangdong Provincial Clinical Research Center for Cancer, State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, China.
| |
Collapse
|
25
|
Huang H, Hu J. Applications of Liquid-Liquid Phase Separation in Biosensing. Chembiochem 2025; 26:e202500028. [PMID: 39920037 DOI: 10.1002/cbic.202500028] [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: 01/13/2025] [Revised: 02/06/2025] [Accepted: 02/07/2025] [Indexed: 02/09/2025]
Abstract
Phase separation, particularly liquid-liquid phase separation (LLPS), has emerged as a powerful tool in biological research, offering unique advantages for visualizing and analyzing biomolecular interactions. This review highlights recent advances in leveraging LLPS to develop experimental techniques for studying protein-protein interactions (PPIs), protein-RNA interactions, and enzyme activity. The integration of LLPS with advanced techniques has expanded its applications, offering new possibilities for unraveling the complexities of cellular function and disease mechanisms. Looking forward, the development of more versatile, sensitive, and targeted LLPS-based methods is poised to transform molecular biology, providing deeper insights into cellular dynamics and facilitating therapeutic advancements.
Collapse
Affiliation(s)
- Huizhen Huang
- Synthetic Biology Center, Haixia Institute of Science and Technology, School of Future Technology, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Jun Hu
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
- Synthetic Biology Center, Haixia Institute of Science and Technology, School of Future Technology, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| |
Collapse
|
26
|
Gounis M, Hamidi H, Ivaska J. mTORC1 shutdown unleashes TFEB to drive triple-negative breast cancer invasion. Dev Cell 2025; 60:979-981. [PMID: 40199239 DOI: 10.1016/j.devcel.2025.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 03/07/2025] [Accepted: 03/07/2025] [Indexed: 04/10/2025]
Abstract
The PI3K/AKT/mTOR pathway is considered a key therapeutic target in triple-negative breast cancer (TNBC). In this issue of Developmental Cell, Remy et al. challenge this idea by demonstrating that mTORC1 inhibition activates TFEB, promoting MT1-MMP exocytosis, ECM degradation, and increased cell invasion, especially when combined with chemotherapy.
Collapse
Affiliation(s)
- Michalis Gounis
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| | - Hellyeh Hamidi
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| | - Johanna Ivaska
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland; Department of Life Technologies, University of Turku, 20520 Turku, Finland; InFLAMES Research Flagship, University of Turku, 20014 Turku, Finland; Western Finnish Cancer Center (FICAN West), University of Turku, 20520 Turku, Finland; Foundation for the Finnish Cancer Institute, Tukholmankatu 8, 00014 Helsinki, Finland.
| |
Collapse
|
27
|
Remy D, Antoine-Bally S, de Toqueville S, Jolly C, Macé AS, Champenois G, Nemati F, Brito I, Raynal V, Priya A, Berlioz A, Dahmani A, Nicolas A, Meseure D, Marangoni E, Chavrier P. TFEB triggers a matrix degradation and invasion program in triple-negative breast cancer cells upon mTORC1 repression. Dev Cell 2025; 60:1018-1035.e8. [PMID: 39729986 DOI: 10.1016/j.devcel.2024.12.005] [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: 11/13/2023] [Revised: 08/14/2024] [Accepted: 12/02/2024] [Indexed: 12/29/2024]
Abstract
The phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) pathway is frequently hyperactivated in triple-negative breast cancers (TNBCs) associated with poor prognosis and is a therapeutic target in breast cancer management. Here, we describe the effects of repression of mTOR-containing complex 1 (mTORC1) through knockdown of several key mTORC1 components or with mTOR inhibitors used in cancer therapy. mTORC1 repression results in an ∼10-fold increase in extracellular matrix proteolytic degradation. Repression in several TNBC models, including in patient-derived xenografts (PDXs), induces nuclear translocation of transcription factor EB (TFEB), which drives a transcriptional program that controls endolysosome function and exocytosis. This response triggers a surge in endolysosomal recycling and the surface exposure of membrane type 1 matrix metalloproteinase (MT1-MMP) associated with invadopodia hyperfunctionality. Furthermore, repression of mTORC1 results in a basal-like breast cancer cell phenotype and disruption of ductal carcinoma in situ (DCIS)-like organization in a tumor xenograft model. Altogether, our data call for revaluation of mTOR inhibitors in breast cancer therapy.
Collapse
Affiliation(s)
- David Remy
- Institut Curie, CNRS UMR 144, PSL University, 75005 Paris, France.
| | | | | | - Célia Jolly
- Institut Curie, CNRS UMR 144, PSL University, 75005 Paris, France
| | - Anne-Sophie Macé
- CurieCoreTech Cell and Tissue Imaging (PICT-IBiSA), Institut Curie, PSL University, 75005 Paris, France
| | | | - Fariba Nemati
- Laboratory of Preclinical Investigation, Institut Curie, PSL University, 26 Rue d'Ulm, 75005 Paris, France
| | - Isabel Brito
- CurieCoreTech Bioinformatics (CUBIC) Platform, Institut Curie, PSL University, 75005 Paris, France
| | - Virginie Raynal
- CurieCoreTech Next Generation Sequencing (ICGex) Platform, Institut Curie, PSL University, 75005 Paris, France
| | - Amulya Priya
- Institut Curie, CNRS UMR 144, PSL University, 75005 Paris, France
| | - Adèle Berlioz
- Institut Curie, CNRS UMR 144, PSL University, 75005 Paris, France
| | - Ahmed Dahmani
- Laboratory of Preclinical Investigation, Institut Curie, PSL University, 26 Rue d'Ulm, 75005 Paris, France
| | - André Nicolas
- Experimental Pathology Platform, Institut Curie, 75005 Paris, France
| | - Didier Meseure
- Experimental Pathology Platform, Institut Curie, 75005 Paris, France
| | - Elisabetta Marangoni
- Laboratory of Preclinical Investigation, Institut Curie, PSL University, 26 Rue d'Ulm, 75005 Paris, France
| | | |
Collapse
|
28
|
Zhang Y, Wang T, Wang Z, Shi X, Jin J. Functions and Therapeutic Potentials of Long Noncoding RNA in Skeletal Muscle Atrophy and Dystrophy. J Cachexia Sarcopenia Muscle 2025; 16:e13747. [PMID: 40034097 PMCID: PMC11876862 DOI: 10.1002/jcsm.13747] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 12/23/2024] [Accepted: 02/04/2025] [Indexed: 03/05/2025] Open
Abstract
Skeletal muscle is the most abundant tissue in the human body and is responsible for movement, metabolism, energy production and longevity. Muscle atrophy is a frequent complication of several diseases and occurs when protein degradation exceeds protein synthesis. Genetics, ageing, nerve injury, weightlessness, cancer, chronic diseases, the accumulation of metabolic byproducts and other stimuli can lead to muscle atrophy. Muscular dystrophy is a neuromuscular disorder, part of which is caused by the deficiency of dystrophin protein and is mostly related to genetics. Muscle atrophy and muscular dystrophy are accompanied by dynamic changes in transcriptomic, translational and epigenetic regulation. Multiple signalling pathways, such as the transforming growth factor-β (TGF-β) signalling pathway, the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/mechanistic target of rapamycin (mTOR) pathway, inflammatory signalling pathways, neuromechanical signalling pathways, endoplasmic reticulum stress and glucocorticoids signalling pathways, regulate muscle atrophy. A large number of long noncoding RNAs (lncRNAs) have been found to be abnormally expressed in atrophic muscles and dystrophic muscles and regulate the balance of muscle protein synthesis and degradation or dystrophin protein expression. These lncRNAs may serve as potential targets for treating muscle atrophy and muscular dystrophy. In this review, we summarized the known lncRNAs related to muscular dystrophy and muscle atrophy induced by denervation, ageing, weightlessness, cachexia and abnormal myogenesis, along with their molecular mechanisms. Finally, we explored the potential of using these lncRNAs as therapeutic targets for muscle atrophy and muscular dystrophy, including the methods of discovery and clinical application prospects for functional lncRNAs.
Collapse
Affiliation(s)
- Yidi Zhang
- Laboratory of Animal Fat Deposition and Muscle Development, Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
| | - Teng Wang
- Laboratory of Animal Fat Deposition and Muscle Development, Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
| | - Ziang Wang
- Laboratory of Animal Fat Deposition and Muscle Development, Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
| | - Xin'e Shi
- Laboratory of Animal Fat Deposition and Muscle Development, Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
| | - Jianjun Jin
- Laboratory of Animal Fat Deposition and Muscle Development, Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
| |
Collapse
|
29
|
Ni M, Zhu Y, Chen Y, Zhao S, Gao A, Lu J, Wang W, Liu R, Gu W, Hong J, Wang J. A gain-of-function variant in RICTOR predisposes to human obesity. J Genet Genomics 2025; 52:549-558. [PMID: 39984155 DOI: 10.1016/j.jgg.2025.02.002] [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: 10/25/2024] [Revised: 02/09/2025] [Accepted: 02/09/2025] [Indexed: 02/23/2025]
Abstract
mTORC1/2 play central roles as signaling hubs of cell growth and metabolism and are therapeutic targets for several diseases. However, the human genetic evidence linking mutations of mTORC1/2 to obesity remains elusive. Using whole-exome sequencing of 1944 cases with severe obesity and 2161 healthy lean controls, we identify a rare RICTOR p.I116V variant enriched in 9 unrelated cases. In Rictor null mouse embryonic fibroblasts, overexpression of the RICTOR p.I116V mutant increases phosphorylation of AKT, a canonical mTORC2 substrate, compared with wild-type RICTOR, indicating a gain-of-function change. Consistent with the human obesity phenotype, the knock-in mice carrying homogenous Rictor p.I116V variants gain more body weight under a high-fat diet. Additionally, the stromal vascular fraction cells derived from inguinal white adipose tissue of knock-in mice display an enhanced capacity for adipocyte differentiation via AKT activity. These findings demonstrate that the rare gain-of-function RICTOR p.I116V mutation activates AKT signaling, promotes adipogenesis, and contributes to obesity in humans.
Collapse
Affiliation(s)
- Mengshan Ni
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai 200025, China
| | - Yinmeng Zhu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai 200025, China
| | - Yufei Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai 200025, China
| | - Shaoqian Zhao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai 200025, China
| | - Aibo Gao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai 200025, China
| | - Jieli Lu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai 200025, China
| | - Weiqing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai 200025, China
| | - Ruixin Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai 200025, China
| | - Weiqiong Gu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai 200025, China.
| | - Jie Hong
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai 200025, China.
| | - Jiqiu Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai 200025, China.
| |
Collapse
|
30
|
Xing Z, Zhang Y, Kang H, Dong H, Zhu D, Liu Y, Sun C, Guo P, Hu B, Tan A. ABHD5 regulates midgut-specific lipid homeostasis in Bombyx mori. INSECT SCIENCE 2025; 32:425-436. [PMID: 38841829 DOI: 10.1111/1744-7917.13386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 03/27/2024] [Accepted: 05/03/2024] [Indexed: 06/07/2024]
Abstract
Lipids are an important energy source and are utilized as substrates for various physiological processes in insects. Comparative gene identification 58 (CGI-58), also known as α/β hydrolase domain-containing 5 (ABHD5), is a highly conserved and multifunctional gene involved in regulating lipid metabolism and cellular energy balance in many organisms. However, the biological functions of ABHD5 in insects are poorly understood. In the current study, we describe the identification and characterization of the ABHD5 gene in the lepidopteran model insect, Bombyx mori. The tissue expression profile investigated using quantitative reverse transcription polymerase chain reaction (RT-qPCR) reveals that BmABHD5 is widely expressed in all tissues, with particularly high levels found in the midgut and testis. A binary transgenic CRISPR/Cas9 system was employed to conduct a functional analysis of BmABHD5, with the mutation of BmABHD5 leading to the dysregulation of lipid metabolism and excessive lipid accumulation in the larval midgut. Histological and physiological analysis further reveals a significant accumulation of lipid droplets in the midgut of mutant larvae. RNA-seq and RT-qPCR analysis showed that genes related to metabolic pathways were significantly affected by the absence of BmABHD5. Altogether, our data prove that BmABHD5 plays an important role in regulating tissue-specific lipid metabolism in the silkworm midgut.
Collapse
Affiliation(s)
- Zhiping Xing
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu Province, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, Jiangsu Province, China
| | - Yuting Zhang
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu Province, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, Jiangsu Province, China
| | - Hongxia Kang
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu Province, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, Jiangsu Province, China
| | - Hui Dong
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu Province, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, Jiangsu Province, China
| | - Dalin Zhu
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu Province, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, Jiangsu Province, China
| | - Yutong Liu
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu Province, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, Jiangsu Province, China
| | - Chenxin Sun
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu Province, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, Jiangsu Province, China
| | - Peilin Guo
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu Province, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, Jiangsu Province, China
| | - Bo Hu
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu Province, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, Jiangsu Province, China
| | - Anjiang Tan
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu Province, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, Jiangsu Province, China
| |
Collapse
|
31
|
Shi XN, Liu CY, Li L, Yao ML, Zhong Z, Jiang YM. The role and therapeutic potential of mitophagy in major depressive disorder. Front Pharmacol 2025; 16:1564276. [PMID: 40206060 PMCID: PMC11979158 DOI: 10.3389/fphar.2025.1564276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 03/05/2025] [Indexed: 04/11/2025] Open
Abstract
Major depressive disorder, also known as MDD, affects more than 264 million people globally, making it a prevalent and critical health challenge. Traditional treatments show limited efficacy in many patients. Therefore, exploring new treatment methods is particularly crucial. Mitophagy, as a regulatory process, can help understand and treat MDD. This paper focuses on the molecular mechanisms of mitophagy, starting from proteins and related pathways, and its role in MDD. The study also explores the associations between mitophagy and neuroinflammation, oxidative stress, neurotransmitter synthesis, and neuroplasticity in MDD and discusses the progress of clinical research on the role of mitophagy in MDD. In addition, the article describes the current pharmaceutical and non-pharmaceutical interventions that can regulate mitophagy in MDD and unravels the potential and challenges of these therapeutic strategies in clinical settings. This article offers a deeper insight into the pathogenesis of MDD and offers a scientific basis for the development of new treatment strategies.
Collapse
Affiliation(s)
- Xin-Nuan Shi
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Chen-Yue Liu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lin Li
- Traditional Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, China
| | - Ming-Li Yao
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Zhen Zhong
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - You-Ming Jiang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
32
|
Li Y, Sun S, Li G, Yang Z, Xing Y, Wang R, Xuan Y, Yang X. The TOR Signaling Pathway Governs Fungal Development, Virulence and Ustiloxin Biosynthesis in Ustilaginoidea virens. J Fungi (Basel) 2025; 11:239. [PMID: 40278060 PMCID: PMC12028740 DOI: 10.3390/jof11040239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/11/2025] [Accepted: 03/20/2025] [Indexed: 04/26/2025] Open
Abstract
Ustilaginoidea virens is an economically important plant pathogen that causes rice false smut, which causes yield reduction and produces mycotoxins in infected grains that pose a serious threat to human and animal health. The target of rapamycin (TOR) signaling pathway acts as a master regular in regulating cell growth and secondary metabolism in fungi. However, little is known about the function of the TOR pathway in regulating fungal development, pathogenicity and mycotoxin biosynthesis in U. virens. Here, we demonstrate that the TOR signaling pathway positively regulates the cell growth, conidiation and pathogenicity in U. virens through the biochemical inhibition of TOR kinases. The inhibition of TOR in U. virens (UvTOR) by rapamycin significantly induces the expression of genes related to mycotoxin biosynthesis, especially that of ustiloxins. Transcriptome analysis under TOR inhibition revealed that the TOR signaling pathway is a regulatory hub that governs U. virens growth and metabolism. A total of 275 differentially expressed genes (DEGs), consisting of 109 up-regulated DEGs and 166 down-regulated DEGs, were identified after rapamycin treatment. The up-regulated DEGs were enriched in amino acid- and acetyl-CoA-related metabolism pathways and the down-regulated DEGs were enriched in carbohydrate- and fatty acid-related metabolism pathways. Collectively, our results provide the first in-depth insight into the TOR signaling pathway in regulating vegetable growth, virulence and mycotoxin biosynthesis in U. virens.
Collapse
Affiliation(s)
- Yuejiao Li
- Institute of Plant Protection, Tianjin Academy of Agricultural Sciences, Tianjin 300381, China; (Y.L.); (S.S.)
| | - Shuqin Sun
- Institute of Plant Protection, Tianjin Academy of Agricultural Sciences, Tianjin 300381, China; (Y.L.); (S.S.)
| | - Guangsheng Li
- Institute of Plant Protection, Tianjin Academy of Agricultural Sciences, Tianjin 300381, China; (Y.L.); (S.S.)
| | - Zezhong Yang
- Institute of Plant Protection, Tianjin Academy of Agricultural Sciences, Tianjin 300381, China; (Y.L.); (S.S.)
| | - Yuqi Xing
- Institute of Plant Protection, Tianjin Academy of Agricultural Sciences, Tianjin 300381, China; (Y.L.); (S.S.)
| | - Ruixiang Wang
- Institute of Plant Protection, Tianjin Academy of Agricultural Sciences, Tianjin 300381, China; (Y.L.); (S.S.)
| | - Yuanhu Xuan
- State Key Laboratory of Elemento-Organic Chemistry and Department of Plant Protection, National Pesticide Engineering Research Center (Tianjin), Nankai University, Tianjin 300071, China
| | - Xiurong Yang
- Institute of Plant Protection, Tianjin Academy of Agricultural Sciences, Tianjin 300381, China; (Y.L.); (S.S.)
| |
Collapse
|
33
|
Adams-Brown SE, Reid KZ. The Central FacilitaTOR: Coordinating Transcription and Translation in Eukaryotes. Int J Mol Sci 2025; 26:2845. [PMID: 40243440 PMCID: PMC11989106 DOI: 10.3390/ijms26072845] [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: 01/31/2025] [Revised: 03/11/2025] [Accepted: 03/17/2025] [Indexed: 04/18/2025] Open
Abstract
One of the biggest challenges to eukaryotic gene expression is coordinating transcription in the nucleus and protein synthesis in the cytoplasm. However, little is known about how these major steps in gene expression are connected. The Target of Rapamycin (TOR) signaling pathway is crucial in connecting these critical phases of gene expression. Highly conserved among eukaryotic cells, TOR regulates growth, metabolism, and cellular equilibrium in response to changes in nutrients, energy levels, and stress conditions. This review examines the extensive role of TOR in gene expression regulation. We highlight how TOR is involved in phosphorylation, remodeling chromatin structure, and managing the factors that facilitate transcription and translation. Furthermore, the critical functions of TOR extend to processing RNA, assembling RNA-protein complexes, and managing their export from the nucleus, demonstrating its wide-reaching impact throughout the cell. Our discussion emphasizes the integral roles of TOR in bridging the processes of transcription and translation and explores how it orchestrates these complex cellular processes.
Collapse
Affiliation(s)
| | - Ke Zhang Reid
- Department of Biology, Wake Forest University, Winston-Salem, NC 27109, USA
| |
Collapse
|
34
|
Harrer DC, Lüke F, Pukrop T, Ghibelli L, Reichle A, Heudobler D. MEPED as salvage therapy for relapsed/refractory Hodgkin's lymphoma incorporating edited non-oncogene addiction: mTOR as a bottleneck. Front Pharmacol 2025; 16:1553331. [PMID: 40183103 PMCID: PMC11965665 DOI: 10.3389/fphar.2025.1553331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 02/24/2025] [Indexed: 04/05/2025] Open
Abstract
Rescue therapies of relapsed/refractory (r/r) Hodgkin's lymphoma (HL) in the third to sixth-line provide major, yet unresolved problems. The MEPED regimen includes nuclear receptor agonists such as pioglitazone and dexamethasone, which counterbalance HL homeostasis, HL stress response inhibitors, everolimus and COX-2 inhibitor, and a stress response inducer, low-dose metronomic treosulfan. CR (six of seven patients) and long-term cCR in patients receiving no consolidating allogeneic stem cell transplantation highlight MEPED as a potent salvage therapy in advanced refractory HL. MEPED edits everolimus activities in such a way that mTORC1 becomes a non-oncogene addiction bottleneck, hence determining long-term therapy outcome. The implications of the therapeutic paradigm shift toward editing of HL tissue, and particularly mTOR addiction, could prove to be profound for clinical practice, both in terms of outcome and treatment tolerability. The long-term results of MEPED treatment indicate the urgent evaluation of the schedule in a multicenter trial for r/r HL.
Collapse
Affiliation(s)
- Dennis Christoph Harrer
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Florian Lüke
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Regensburg, Germany
- Division of Personalized Tumor Therapy, Fraunhofer Institute for Toxicology and Experimental Medicine, Regensburg, Germany
| | - Tobias Pukrop
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), University Hospital Regensburg, Regensburg, Germany
| | - Lina Ghibelli
- Department of Biology, University of Rome “Tor Vergata”, Rome, Italy
| | - Albrecht Reichle
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Daniel Heudobler
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
35
|
Jacinto E. Making sense of fat in cancer. Science 2025; 387:1147-1148. [PMID: 40080596 DOI: 10.1126/science.adw1956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
A lipid chaperone enables sensing of an essential fatty acid to drive tumor growth.
Collapse
Affiliation(s)
- Estela Jacinto
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA
| |
Collapse
|
36
|
Hu Y, Wang K, Xu J, Wan G, Zhao Y, Chen Y, Jiang K, Li X. mTOR-Mediated Autophagy Regulates Cadmium-Induced Kidney Injury via Pyroptosis. Int J Mol Sci 2025; 26:2589. [PMID: 40141229 PMCID: PMC11942160 DOI: 10.3390/ijms26062589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 02/17/2025] [Accepted: 02/22/2025] [Indexed: 03/28/2025] Open
Abstract
The heavy metal cadmium (Cd) affects the global livestock production economy mainly through the contamination of feed raw materials and secondary contamination in feed processing, and it also poses a serious threat to food safety and human health. The nucleotide-binding oligomerization domain-like pyrin-domain-containing protein 3 (NLRP3) inflammasome is a key regulatory element of pyroptosis, which is engaged in kidney injury. Meanwhile, autophagy is also involved in renal inflammation. Mammalian target of rapamycin (mTOR) plays an important role in pyroptosis and autophagy, but its function in Cd-induced kidney injury remains unclear. In this study, we explored the role of mTOR-mediated autophagy and pyroptosis in kidney injury caused by Cd exposure and elucidated its underlying mechanism. Our data showed that Cd exposure reduced the integrity of kidney cell membranes, increased the expression of pyroptosis-associated proteins, and promoted the release of inflammatory cytokines. Subsequently, a notable attenuation in Cd-induced pyroptosis was observed following the administration of CY-09, an NLRP3 inhibitor. In addition, Cd exposure promoted autophagy in kidney cells. Importantly, in both in vivo and in vitro experiments, rapamycin, an mTOR inhibitor, downregulated the expression of pyroptosis-related proteins, thereby significantly improving Cd-induced kidney injury. In summary, our results indicate that mTOR-mediated autophagy has a significant protective effect on NLRP3 inflammasome-dependent kidney injury induced by Cd exposure, thus providing new insights into the prevention and treatment of Cd poisoning.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Kangfeng Jiang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (Y.H.); (K.W.); (J.X.); (G.W.); (Y.Z.); (Y.C.)
| | - Xiaobing Li
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China; (Y.H.); (K.W.); (J.X.); (G.W.); (Y.Z.); (Y.C.)
| |
Collapse
|
37
|
Ryspayeva D, Seyhan AA, MacDonald WJ, Purcell C, Roady TJ, Ghandali M, Verovkina N, El-Deiry WS, Taylor MS, Graff SL. Signaling pathway dysregulation in breast cancer. Oncotarget 2025; 16:168-201. [PMID: 40080721 PMCID: PMC11906143 DOI: 10.18632/oncotarget.28701] [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: 12/20/2024] [Accepted: 03/03/2025] [Indexed: 03/15/2025] Open
Abstract
This article provides a comprehensive analysis of the signaling pathways implicated in breast cancer (BC), the most prevalent malignancy among women and a leading cause of cancer-related mortality globally. Special emphasis is placed on the structural dynamics of protein complexes that are integral to the regulation of these signaling cascades. Dysregulation of cellular signaling is a fundamental aspect of BC pathophysiology, with both upstream and downstream signaling cascade activation contributing to cellular process aberrations that not only drive tumor growth, but also contribute to resistance against current treatments. The review explores alterations within these pathways across different BC subtypes and highlights potential therapeutic strategies targeting these pathways. Additionally, the influence of specific mutations on therapeutic decision-making is examined, underscoring their relevance to particular BC subtypes. The article also discusses both approved therapeutic modalities and ongoing clinical trials targeting disrupted signaling pathways. However, further investigation is necessary to fully elucidate the underlying mechanisms and optimize personalized treatment approaches.
Collapse
Affiliation(s)
- Dinara Ryspayeva
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
| | - Attila A. Seyhan
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
- Pathobiology Graduate Program, Brown University, RI 02903, USA
| | - William J. MacDonald
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
| | - Connor Purcell
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
| | - Tyler J. Roady
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
- Pathobiology Graduate Program, Brown University, RI 02903, USA
| | - Maryam Ghandali
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
| | - Nataliia Verovkina
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
| | - Wafik S. El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
- Pathobiology Graduate Program, Brown University, RI 02903, USA
- Department of Medicine, Hematology/Oncology Division, Lifespan Health System and Brown University, RI 02903, USA
| | - Martin S. Taylor
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
- Pathobiology Graduate Program, Brown University, RI 02903, USA
- Brown Center on the Biology of Aging, Brown University, RI 02903, USA
| | - Stephanie L. Graff
- Legorreta Cancer Center at Brown University, RI 02903, USA
- Department of Medicine, Hematology/Oncology Division, Lifespan Health System and Brown University, RI 02903, USA
| |
Collapse
|
38
|
Zhou W, Sheng Y, Hu D, An Y, Yang M, Wang W, Basnet S, Yan J, Zhang S, Liu Q, Li Y, Tan Y, Gao J, Sun K, Du C. Proteasome inhibition induces DNA methylation alteration by attenuating the synthesis of DNA methyltransferase 1 and 3B in colorectal cancer. Sci Rep 2025; 15:8534. [PMID: 40075132 PMCID: PMC11903776 DOI: 10.1038/s41598-025-92390-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
Proteasome is an essential organelle responsible for maintaining cellular protein homeostasis, but its relationship with DNA methylation remains unknown. In this study, we assessed DNA methylation of colorectal cancer (CRC) cells following treatment with proteasome inhibitors, and investigated the underlying mechanism of DNA methylation changes and the biological effects on CRC cells. We established that inhibition of proteasome leads to significant alterations in DNA methylation profile in CRC by suppressing the synthesis of DNA methyltransferases (DNMTs). We found that treating CRC cells with proteasome inhibitors results in attenuated translation of DNMT1 and DNMT3B, mediated by the inactivation of AKT and mammalian target of rapamycin (mTOR), which is dependent on the accumulation of p300, an acetyltransferase that inhibits AKT through acetylation modification. Furthermore, we demonstrated that downregulation of DNMT1 and DNMT3B confers protection against proteasome inhibitor treatment, potentially through reprogramming the transcriptome of CRC cells, highlighting the significant role of DNMTs in response to disruptions in protein homeostasis. Interestingly, it appears that the proteasome inhibitor-induced downregulation of DNMT1 and DNMT3B is specific to CRC. Altogether, our findings reveal an epigenetic effect of proteasome on DNA methylation in CRC through its regulation of DNA methyltransferase synthesis.
Collapse
Affiliation(s)
- Wenwen Zhou
- Key University Laboratory of Metabolism and Health of Guangdong, Biochemistry Department, School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Avenue, Shenzhen, 518055, Guangdong, People's Republic of China
| | - Yuling Sheng
- Key University Laboratory of Metabolism and Health of Guangdong, Biochemistry Department, School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Avenue, Shenzhen, 518055, Guangdong, People's Republic of China
| | - Dingxue Hu
- Institute of Cancer Research, Shenzhen Bay Laboratory, Gaoke Innovation Center, Room A1921, Guangming District, Shenzhen, 518132, People's Republic of China
| | - Yunyun An
- Institute of Cancer Research, Shenzhen Bay Laboratory, Gaoke Innovation Center, Room A1921, Guangming District, Shenzhen, 518132, People's Republic of China
| | - Mengqi Yang
- Institute of Cancer Research, Shenzhen Bay Laboratory, Gaoke Innovation Center, Room A1921, Guangming District, Shenzhen, 518132, People's Republic of China
| | - Wanqiu Wang
- Institute of Cancer Research, Shenzhen Bay Laboratory, Gaoke Innovation Center, Room A1921, Guangming District, Shenzhen, 518132, People's Republic of China
| | - Shiva Basnet
- Key University Laboratory of Metabolism and Health of Guangdong, Biochemistry Department, School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Avenue, Shenzhen, 518055, Guangdong, People's Republic of China
| | - Jingyu Yan
- Key University Laboratory of Metabolism and Health of Guangdong, Biochemistry Department, School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Avenue, Shenzhen, 518055, Guangdong, People's Republic of China
| | - Shuxia Zhang
- Key University Laboratory of Metabolism and Health of Guangdong, Biochemistry Department, School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Avenue, Shenzhen, 518055, Guangdong, People's Republic of China
| | - Qi Liu
- Key University Laboratory of Metabolism and Health of Guangdong, Biochemistry Department, School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Avenue, Shenzhen, 518055, Guangdong, People's Republic of China
| | - Yunze Li
- Key University Laboratory of Metabolism and Health of Guangdong, Biochemistry Department, School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Avenue, Shenzhen, 518055, Guangdong, People's Republic of China
| | - Yi Tan
- Key University Laboratory of Metabolism and Health of Guangdong, Biochemistry Department, School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Avenue, Shenzhen, 518055, Guangdong, People's Republic of China
| | - Jing Gao
- Peking University Shenzhen Hospital, 1120 Lianhua Road, Shenzhen, 516473, Guangdong, People's Republic of China
| | - Kun Sun
- Institute of Cancer Research, Shenzhen Bay Laboratory, Gaoke Innovation Center, Room A1921, Guangming District, Shenzhen, 518132, People's Republic of China.
| | - Changzheng Du
- Cancer Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine, Tsinghua University, 168 Litang Road, Changping District, Beijing, 102218, People's Republic of China.
- School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Avenue, Shenzhen, 518055, Guangdong, People's Republic of China.
| |
Collapse
|
39
|
Singh G, Rohit, Kumar P, Aran KR. Targeting EGFR and PI3K/mTOR pathways in glioblastoma: innovative therapeutic approaches. Med Oncol 2025; 42:97. [PMID: 40064710 DOI: 10.1007/s12032-025-02652-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 02/24/2025] [Indexed: 03/29/2025]
Abstract
Glioblastoma (GBM) stands as the most aggressive form of primary brain cancer in adults, characterized by its rapid growth, invasive nature, and a robust propensity to induce angiogenesis, forming new blood vessels to sustain its expansion. GBM arises from astrocytes, star-shaped glial cells, and despite significant progress in understanding its molecular mechanisms, its prognosis remains grim. It is frequently associated with mutations or overexpression of the epidermal growth factor receptor (EGFR), which initiates several downstream signaling pathways. Dysregulation of key signaling pathways, such as EGFR/PTEN/AKT/mTOR, drives tumorigenesis, promotes metastasis and leads to treatment resistance. The modest survival benefits of the conventional treatment of surgical resection followed by radiation and chemotherapy underscore the pressing need for innovative therapeutic approaches. In most the tumor, overexpression of EGFR is found associated with GBM and mutations in its several variants are important for promoting ongoing mitogenic signaling and tumor growth. This receptor inhibits apoptosis and promotes cell survival and proliferation by activating downstream PI3K/AKT/mTOR pathways. This route is typically blocked by PTEN, a crucial tumor suppressor, however, GBM frequently results in abnormalities in this protein. The aim of this review is to explore the molecular foundations of GBM, with a focus on the EGFR and PI3K/mTOR pathways and their impact on tumor behavior. Additionally, this review highlights EGFR and PI3K/AKT/mTOR inhibitors currently in clinical and preclinical trials, addressing treatment resistance, challenges, and future directions.
Collapse
Affiliation(s)
- Gursimran Singh
- Department of Pharmacy Practice, ISF College of Pharmacy (an Autonomous College), Moga, Punjab, 142001, India
| | - Rohit
- Research Scholar, I.K. Gujral Punjab Technical University, Kapurthala, Punjab, 144603, India
| | - Pankaj Kumar
- Department of Pharmacology, Himachal Institute of Pharmaceutical Education and Research (HIPER), Tehsil-Nadaun, Hamirpur, Himachal Pradesh, 177033, India
| | - Khadga Raj Aran
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy (an Autonomous College), Moga, Punjab, 142001, India.
| |
Collapse
|
40
|
Yu P, Zhao X, Zhou D, Wang S, Hu Z, Lian K, Zhang N, Duan P. The microRNA-mediated apoptotic signaling axis in male reproduction: a possible and targetable culprit in male infertility. Cell Biol Toxicol 2025; 41:54. [PMID: 40038116 PMCID: PMC11880093 DOI: 10.1007/s10565-025-10006-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 02/20/2025] [Indexed: 03/06/2025]
Abstract
Recently, infertility has emerged as a significant and prevalent public health concern warranting considerable attention. Apoptosis, recognized as programmed cell death, constitutes a crucial process essential for the maintenance of normal spermatogenesis. Multiple investigations have illustrated that the dysregulated apoptosis of reproductive cells, encompassing spermatogonial stem cells, Sertoli cells, and Leydig cells, serves as a causative factor in male infertility. MicroRNAs represent a class of small RNA molecules that exert negative regulatory control over gene expression using direct interaction with messenger RNA transcripts. Previous studies have established that aberrant expression of miRNAs induces apoptosis in reproductive tissues, correlating with reproductive dysfunctions and infertility. In this review, we offer a comprehensive overview of miRNAs and their respective target genes implicated in the apoptotic process. As well, miRNAs are involved in multiple apoptotic signaling pathways, namely the PI3K/AKT, NOTCH, Wnt/β-catenin, and mTOR signaling cascades, exerting both negative and positive effects. We additionally elucidate the significant functions played by lncRNAs and circular RNAs as competing endogenous RNAs in the process of apoptosis within reproductive cells. We further illustrate that external factors, including silica nanoparticles, Cyclosporine A, and smoking, induce dysregulation of miRNAs, resulting in apoptosis within reproductive cells and subsequent male reproductive toxicity. Further, we discuss the implication of heat stress, hypoxia, and diabetes in reproductive cell apoptosis induced by miRNA dysregulation in male infertility. Finally, we demonstrate that the modulation of miRNAs via traditional and novel medicine could protect reproductive cells from apoptosis and be implemented as a therapeutic approach in male infertility.
Collapse
Affiliation(s)
- Pengxia Yu
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Department of Obstetrics and Gynecology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, 441000, China
- Hubei Provincial Clinical Research Center for Accurate Fetus Malformation Diagnosis, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, 441000, China
| | - Xue Zhao
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Department of Obstetrics and Gynecology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, 441000, China
- Department of Pharmacology, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, China
| | - Dan Zhou
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Department of Obstetrics and Gynecology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, 441000, China
| | - Songtao Wang
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Department of Obstetrics and Gynecology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, 441000, China
| | - Zihuan Hu
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Department of Obstetrics and Gynecology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, 441000, China
| | - Kai Lian
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Department of Obstetrics and Gynecology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, 441000, China
| | - Nanhui Zhang
- Department of Nephrology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, 441000, China.
| | - Peng Duan
- Key Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases of Xiangyang City, Department of Obstetrics and Gynecology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, 441000, China.
- Hubei Provincial Clinical Research Center for Accurate Fetus Malformation Diagnosis, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, 441000, China.
| |
Collapse
|
41
|
Song Y, Wang F, Luo H, Hu H, Pang Y, Xu K, Zhang X. Rapamycin protects glucocorticoid-induced glaucoma model mice against trabecular meshwork fibrosis by suppressing mTORC1/2 signaling. Eur J Pharmacol 2025; 990:177269. [PMID: 39805488 DOI: 10.1016/j.ejphar.2025.177269] [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: 08/07/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 01/16/2025]
Abstract
Systemic or local use of glucocorticoids (GCs) can induce pathological elevation of intraocular pressure (IOP), potentially leading to permanent visual loss. Previous studies have demonstrated that rapamycin (Rapa) inhibits the activation of retinal glial cells (RGC) and the production of neuroinflammation, achieving neuroprotective goals. However, there has been little research on the effect of Rapa on the trabecular meshwork (TM). This study aimed to investigate the protective effect and potential mechanism of Rapa in a glucocorticoid-induced glaucoma (GIG) model. Our findings indicate that Rapa significantly inhibited the IOP increase induced by dexamethasone acetate (Dex-Ac) and improved TM fibrosis and RGC damage. In cultured human trabecular meshwork cells (HTMCs) treated with dexamethasone (Dex) and Rapa under different conditions revealed that Rapa inhibits Dex-induced HTMC fibrosis and cytoskeletal changes. This effect may result from the specific suppression of the mechanistic target of rapamycin complex 1 (mTORC1) pathway by Rapa, which reduces abnormal extracellular matrix (ECM) deposition. Alternatively, the improvement in cytoskeleton entanglement might be due to the inhibition of the mechanistic target of rapamycin complex 2 (mTORC2) pathway. These two potential mechanisms may collectively contribute to the protective effects of Rapa in GIG. This study provides a new theoretical basis for using of Rapa in the treatment of GIG.
Collapse
Affiliation(s)
- Yuning Song
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology & Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, Nanchang, China
| | - Feifei Wang
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology & Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, Nanchang, China; Department of Ophthalmology, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Hongdou Luo
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology & Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, Nanchang, China
| | - Haijian Hu
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology & Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, Nanchang, China
| | - Yulian Pang
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology & Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, Nanchang, China
| | - Ke Xu
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology & Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, Nanchang, China
| | - Xu Zhang
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology & Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, Nanchang, China.
| |
Collapse
|
42
|
Huang Y, Gao Y, Lin Z, Miao H. Involvement of the ubiquitin-proteasome system in the regulation of the tumor microenvironment and progression. Genes Dis 2025; 12:101240. [PMID: 39759114 PMCID: PMC11697063 DOI: 10.1016/j.gendis.2024.101240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 10/11/2023] [Accepted: 12/05/2023] [Indexed: 01/07/2025] Open
Abstract
The tumor microenvironment is a complex environment comprising tumor cells, non-tumor cells, and other critical non-cellular components. Some studies about tumor microenvironment have recently achieved remarkable progress in tumor treatment. As a substantial part of post-translational protein modification, ubiquitination is a crucial player in maintaining protein stability in cell signaling, cell growth, and a series of cellular life activities, which are also essential for regulating tumor cells or other non-tumor cells in the tumor microenvironment. This review focuses on the role and function of ubiquitination and deubiquitination modification in the tumor microenvironment while discussing the prospect of developing inhibitors targeting ubiquity-related enzymes, thereby providing ideas for future research in cancer therapy.
Collapse
Affiliation(s)
- Yulan Huang
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing 400038, China
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Yuan Gao
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing 400038, China
| | - Zhenghong Lin
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Hongming Miao
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing 400038, China
- School of Life Sciences, Chongqing University, Chongqing 401331, China
- Jinfeng Laboratory, Chongqing 401329, China
| |
Collapse
|
43
|
James NR, O'Neill JS. Circadian Control of Protein Synthesis. Bioessays 2025; 47:e202300158. [PMID: 39668398 PMCID: PMC11848126 DOI: 10.1002/bies.202300158] [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: 08/22/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 12/14/2024]
Abstract
Daily rhythms in the rate and specificity of protein synthesis occur in most mammalian cells through an interaction between cell-autonomous circadian regulation and daily cycles of systemic cues. However, the overall protein content of a typical cell changes little over 24 h. For most proteins, translation appears to be coordinated with protein degradation, producing phases of proteomic renewal that maximize energy efficiency while broadly maintaining proteostasis across the solar cycle. We propose that a major function of this temporal compartmentalization-and of circadian rhythmicity in general-is to optimize the energy efficiency of protein synthesis and associated processes such as complex assembly. We further propose that much of this temporal compartmentalization is achieved at the level of translational initiation, such that the translational machinery alternates between distinct translational mechanisms, each using a distinct toolkit of phosphoproteins to preferentially recognize and translate different classes of mRNA.
Collapse
Affiliation(s)
- Nathan R. James
- Division of Cell BiologyMRC Laboratory of Molecular BiologyCambridgeUK
| | - John S. O'Neill
- Division of Cell BiologyMRC Laboratory of Molecular BiologyCambridgeUK
| |
Collapse
|
44
|
Lanuza-Gracia P, Juan-Mateu J, Valcárcel J. Splice age: mTORC1-mediated RNA splicing in metabolism and ageing. Trends Cell Biol 2025; 35:183-185. [PMID: 39843329 DOI: 10.1016/j.tcb.2025.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/03/2025] [Accepted: 01/06/2025] [Indexed: 01/24/2025]
Abstract
The target of rapamycin complex mTORC1 has key roles in cell growth and metabolism and its inhibition delays ageing. Recent work by Ogawa et al. in Caenorhabditis elegans argues that modulation of pre-mRNA splicing factors and alternative splicing are key mediators of mTORC1 signalling and can enhance longevity.
Collapse
Affiliation(s)
- Pablo Lanuza-Gracia
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr Aiguader 88, 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), Dr Aiguader 88, 08003, Barcelona, Spain
| | - Jonas Juan-Mateu
- Universitat Pompeu Fabra (UPF), Dr Aiguader 88, 08003, Barcelona, Spain
| | - Juan Valcárcel
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr Aiguader 88, 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), Dr Aiguader 88, 08003, Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Passeig Lluis Companys 23, 08010 Barcelona, Spain.
| |
Collapse
|
45
|
Hao Y, Li Z, Du X, Xie Q, Li D, Lei S, Guo Y. Characterization and chemoproteomic profiling of protein O-GlcNAcylation in SOD1-G93A mouse model. Mol Med 2025; 31:82. [PMID: 40021952 PMCID: PMC11871760 DOI: 10.1186/s10020-025-01134-4] [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: 12/14/2024] [Accepted: 02/15/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a devastating motor neuron disease. Protein O-linked β-N-acetylglucosamine (O-GlcNAc) modification has been found to affect the processing of several important proteins implicated in ALS. However, the overall level and cellular localization of O-GlcNAc during ALS progression are incompletely understood, and large-scale profiling of O-GlcNAcylation sites in this context remains unexplored. METHODS By using immunostaining analysis and chemoenzymatic labeling-based quantitative chemoproteomics, we assayed O-GlcNAcylation dynamics of lumbar spinal cords from SOD-G93A mice and their non-transgenic (NTG) littermates, the most widely used animal model for studying ALS pathogenesis. RESULTS We discovered that the global O-GlcNAcylation was significantly reduced at the disease end stage. Correlatively, a great increase of OGA was observed. Immunohistochemistry and immunofluorescence analysis showed a higher proportion of O-GlcNAc-positive neurons in the NTG group, while O-GlcNAc colocalization with astrocytes/microglia was elevated in SOD1-G93A mice. Moreover, we reported the identification of 568 high-confidence O-GlcNAc sites from end-stage SOD1-G93A and NTG mice. Of the 568 sites, 226-many of which occurred on neuronal function and structure-related proteins-were found to be dynamically regulated. CONCLUSION These data provide a valuable resource for dissecting the functional role of O-GlcNAcylation in ALS and shed light on promising therapeutic avenues for ALS. The chemoenzymatic labeling-based chemoproteomic approach is applicable for probing O-GlcNAc dynamics in various pathological processes.
Collapse
Affiliation(s)
- Yi Hao
- National Glycoengineering Research Center, Shandong University, Qingdao, Shandong, China
| | - Zhongzhong Li
- Beijing Geriatric Healthcare and Disease Prevention Center, Xuanwu Hospital, Capital Medical University, Changchun Street 45, Beijing, China
| | - Xinyan Du
- Beijing Geriatric Healthcare and Disease Prevention Center, Xuanwu Hospital, Capital Medical University, Changchun Street 45, Beijing, China
| | - Qingsong Xie
- National Glycoengineering Research Center, Shandong University, Qingdao, Shandong, China
| | - Dongxiao Li
- Department of Neurology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Shaoyuan Lei
- Evidence-Based Medicine Center, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yansu Guo
- Beijing Geriatric Healthcare and Disease Prevention Center, Xuanwu Hospital, Capital Medical University, Changchun Street 45, Beijing, China.
- Evidence-Based Medicine Center, Xuanwu Hospital, Capital Medical University, Beijing, China.
- Beijing Municipal Geriatric Medical Research Center, Beijing, China.
| |
Collapse
|
46
|
Jefcoate CR, Larsen MC, Song YS, Maguire M, Sheibani N. Defined Diets Link Iron and α-Linolenic Acid to Cyp1b1 Regulation of Neonatal Liver Development Through Srebp Forms and LncRNA H19. Int J Mol Sci 2025; 26:2011. [PMID: 40076634 PMCID: PMC11901102 DOI: 10.3390/ijms26052011] [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: 11/02/2024] [Revised: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 03/14/2025] Open
Abstract
Cyp1b1 substantially affects hepatic vascular and stellate cells (HSC) with linkage to liver fibrosis. Despite minimal hepatocyte expression, Cyp1b1 deletion substantially impacts liver gene expression at birth and weaning. The appreciable Cyp1b1 expression in surrounding embryo mesenchyme, during early organogenesis, provides a likely source for Cyp1b1. Here defined breeder diets established major interconnected effects on neonatal liver of α-linolenic acid (ALA), vitamin A deficiency (VAD) and suboptimal iron fed mice. At birth Cyp1b1 deletion and VAD each activated perinatal HSC, while suppressing iron control by hepcidin. Cyp1b1 deletion also advanced the expression of diverse genes linked to iron regulation. Postnatal stimulations of Srebp-regulated genes in the fatty acid and cholesterol biosynthesis pathways were suppressed by Cyp1b1-deficiency. LncRNA H19 and the neutrophil alarmin S100a9 expression increased due to slower postnatal decline with Cyp1b1 deficiency. VAD reversed each of Cyp1b1 effect, probably due to enhanced HSC release of Apo-Rbp4. At birth, Cyp1b1 deletion enhanced H19 participation. Notably, a suppressor (Cnot3) decreased while an activity partner (Ezh2/H3K methylation) increased H19 expression. ALA elevated hepcidin mRNA and countered the inhibitory effects of Cyp1b1 deletion on hepcidin expression. Oxylipin metabolites of ALA from highly expressed hepatic Cyps are potential mediators. Cyp expression patterns demonstrated female dimorphism for neonatal liver. Mothers followed one of three fetal growth support programs probably linked to maturity at conception.
Collapse
Affiliation(s)
- Colin R. Jefcoate
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (M.C.L.); (M.M.)
| | - Michele C. Larsen
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (M.C.L.); (M.M.)
| | - Yong-Seok Song
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA;
| | - Meghan Maguire
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (M.C.L.); (M.M.)
| | - Nader Sheibani
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (M.C.L.); (M.M.)
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA;
| |
Collapse
|
47
|
Klauer MJ, Hall KL, Jagla CAD, Tsvetanova NG. Extensive location bias of the GPCR-dependent translatome via site-selective activation of mTOR. Proc Natl Acad Sci U S A 2025; 122:e2414738122. [PMID: 39964727 PMCID: PMC11874449 DOI: 10.1073/pnas.2414738122] [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: 07/23/2024] [Accepted: 01/13/2025] [Indexed: 02/20/2025] Open
Abstract
G protein-coupled receptors (GPCRs) modulate various physiological functions by rewiring cellular gene expression in response to extracellular signals. Control of gene expression by GPCRs has been studied almost exclusively at the transcriptional level, neglecting an extensive amount of regulation that takes place translationally. Hence, little is known about the nature and mechanisms of gene-specific posttranscriptional regulation downstream of receptor activation. Here, we apply an unbiased multiomics approach to delineate an extensive translational regulatory program initiated by the prototypical beta2-adrenergic receptor (β2-AR) and provide mechanistic insights into how these processes are orchestrated. Using ribosome profiling (Ribo-seq), we identify nearly 120 gene targets of adrenergic receptor activity for which expression is exclusively regulated at the level of translation. We next show that all translational changes are induced selectively by endosomal β2-ARs and report that this proceeds through activation of the mammalian target of rapamycin (mTOR) pathway. Specifically, within the set of translational GPCR targets, we find significant enrichment of genes with 5' terminal oligopyrimidine (TOP) motifs, a gene class classically known to be translationally regulated by mTOR. We then demonstrate that endosomal β2-ARs are required for mTOR activation and subsequent mTOR-dependent TOP mRNA translation. This site-selective crosstalk between the pathways is observed in multiple cell models with native β2-ARs, across a range of endogenous and synthetic adrenergic agonists, and for other GPCRs with intracellular activity. Together, this comprehensive analysis of drug-induced translational regulation establishes a critical role for location-biased GPCR signaling in fine-tuning the cellular protein landscape.
Collapse
Affiliation(s)
- Matthew J. Klauer
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC27710
| | - Katherine L. Hall
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC27710
| | - Caitlin A. D. Jagla
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC27710
| | | |
Collapse
|
48
|
Fang Y, Zhang Y, Huang T, Yang S, Li Y, Zhou L. Focal cortical dysplasia type II: review of neuropathological manifestations and pathogenetic mechanisms. ACTA EPILEPTOLOGICA 2025; 7:12. [PMID: 40217346 PMCID: PMC11960379 DOI: 10.1186/s42494-024-00195-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 11/22/2024] [Indexed: 04/15/2025] Open
Abstract
Focal cortical dysplasia (FCD) is an important cause of intractable epilepsy, with FCD type II (FCD II) being the most common subtype. FCD II is characterized by cortical dyslamination accompanied by dysmorphic neurons (DNs). Identifying the molecular alterations and targetable biomarkers is pivotal for developing therapies. Here, we provide a detailed description of the neuropathological manifestations of FCD II, including morphological alterations and immunophenotypic profiles, indicating that abnormal cells exhibit a diverse spectrum of mixed differentiation states. Furthermore, we summarize current research on the pathogenetic mechanisms, indicating that gene mutations, epigenetic alterations, cortical developmental protein disturbances, inflammatory processes, and extrinsic damages may lead to abnormal neuronal proliferation and migration, thereby contributing to the emergence and progression of FCD II. These findings not only enhance our understanding of the pathogenesis of FCD II but also offer new directions for clinical diagnosis and treatment. Future research should further explore the interactions among these factors and employ multidisciplinary approaches to advance our understanding of FCD II.
Collapse
Affiliation(s)
- Yubao Fang
- Department of Neurology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yaqian Zhang
- Department of Neurology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Tiancai Huang
- Department of Neurology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Shengyu Yang
- Department of Neurology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yinchao Li
- Department of Neurology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Liemin Zhou
- Department of Neurology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China.
| |
Collapse
|
49
|
D'Amore A, Sundberg M, Lin R, Lubbers ET, Winden KD, Yu L, Gawlinska K, Gawlinski D, Lopez SG, Choe Y, Wightman EV, Liang Y, Modi M, Yuskaitis CJ, Lee HHC, Rotenberg A, Sahin M. Phenotypic rescue via mTOR inhibition in neuron-specific Pten knockout mice reveals AKT and mTORC1-site specific changes. Mol Psychiatry 2025:10.1038/s41380-025-02916-2. [PMID: 39953287 DOI: 10.1038/s41380-025-02916-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/22/2024] [Accepted: 01/30/2025] [Indexed: 02/17/2025]
Abstract
Phosphatase and Tensin Homolog (PTEN) is a dual-specific protein and lipid phosphatase that regulates AKT and downstream signaling of the mechanistic target of rapamycin (mTOR). PTEN functions as a tumor suppressor gene whose mutations result in PTEN Hamartoma Tumor Syndrome (PHTS) characterized by increased cancer risk and neurodevelopmental comorbidity. Here, we generated a novel neuron-specific Pten knock-out mouse model (Syn-Cre/Pten HOM) to test the ability of pharmacologic mTOR inhibition to rescue Pten mutation-associated disease phenotypes in vivo and in vitro. We found that treatment with the mTOR inhibitor, everolimus, increased the survival of Syn-Cre/Pten HOM mice while some neurologic phenotypes persisted. Transcriptomic analyses revealed that in contrast to mice harboring a neuron-specific deletion of the Tuberous Sclerosis Complex 2 gene (Syn-Cre/Tsc2 KO), genes that are under AKT regulation were significantly increased in the Syn-Cre/Pten HOM mice. In addition, genes associated with synapse, extracellular matrix, and myelination were broadly increased in Syn-Cre/Pten HOM mouse neocortex. These findings were confirmed by immunostaining of cortical sections in vivo, which revealed excessive immunoreactivity of myelin basic protein and perineuronal nets (PNN), the specialized extracellular matrix surrounding fast-spiking parvalbumin (PV) interneurons. We also detected increased expression of Synapsin I/PSD95 positive synapses and network hyperactivity phenotypes in Syn-Cre/Pten HOM mice neurons compared to wild-type (WT) neurons in vitro. Strikingly, everolimus treatment rescued the number of synapses and network hyperactivity in the Syn-Cre/Pten HOM mice cortical neuron cultures. Taken together, our results revealed in vivo and in vitro molecular and neuronal network mechanisms underlying neurological phenotypes of PHTS. Notably, pharmacologic mTOR inhibition by everolimus led to successful downstream signaling rescue, including mTOR complex 1 (mTORC1) site-specific suppression of S6 phosphorylation, correlating with phenotypic rescue found in our novel neuron-specific Syn-Cre/Pten HOM mice.
Collapse
Affiliation(s)
- Angelica D'Amore
- Department of Neurology, FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, USA
| | - Maria Sundberg
- Department of Neurology, FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, USA
| | - Rui Lin
- Department of Neurology, FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, USA
| | - Ella T Lubbers
- Department of Neurology, FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, USA
| | - Kellen D Winden
- Department of Neurology, FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, USA
| | - Lucy Yu
- Department of Neurology, FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, USA
| | - Kinga Gawlinska
- Department of Neurology, FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, USA
- Department of Clinical Pharmacy, Jagiellonian University, Medical College, Medyczna 9, PL 30-688, Krakow, Poland
| | - Dawid Gawlinski
- Department of Neurology, FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, USA
| | - Sam G Lopez
- Department of Neurology, FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, USA
| | - Yongho Choe
- Department of Neurology, FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, USA
| | - Emma V Wightman
- Department of Neurology, FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, USA
| | - Yini Liang
- Department of Neurology, FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, USA
| | - Meera Modi
- Department of Neurology, FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, USA
| | - Christopher J Yuskaitis
- Department of Neurology, FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, USA
- Division of Epilepsy and Clinical Neurophysiology, Boston Children's Hospital, Boston, USA
| | - Henry Hing Cheong Lee
- Department of Neurology, FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, USA
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, USA
| | - Alexander Rotenberg
- Department of Neurology, FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, USA
- Division of Epilepsy and Clinical Neurophysiology, Boston Children's Hospital, Boston, USA
| | - Mustafa Sahin
- Department of Neurology, FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, USA.
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, USA.
| |
Collapse
|
50
|
Rivera-Correa J, Gupta S, Ricker E, Flores-Castro D, Jenkins D, Vulcano S, Phalke SP, Pannellini T, Miele MM, Li Z, Zamponi N, Kim YB, Chinenov Y, Giannopoulou E, Cerchietti L, Pernis AB. ROCK1 promotes B cell differentiation and proteostasis under stress through the heme-regulated proteins, BACH2 and HRI. JCI Insight 2025; 10:e180507. [PMID: 39903532 PMCID: PMC11949073 DOI: 10.1172/jci.insight.180507] [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: 03/11/2024] [Accepted: 01/28/2025] [Indexed: 02/06/2025] Open
Abstract
The mechanisms utilized by differentiating B cells to withstand highly damaging conditions generated during severe infections, like the massive hemolysis that accompanies malaria, are poorly understood. Here, we demonstrate that ROCK1 regulates B cell differentiation in hostile environments replete with pathogen-associated molecular patterns (PAMPs) and high levels of heme by controlling 2 key heme-regulated molecules, BACH2 and heme-regulated eIF2α kinase (HRI). ROCK1 phosphorylates BACH2 and protects it from heme-driven degradation. As B cells differentiate, furthermore, ROCK1 restrains their pro-inflammatory potential and helps them handle the heightened stress imparted by the presence of PAMPs and heme by controlling HRI, a key regulator of the integrated stress response and cytosolic proteotoxicity. ROCK1 controls the interplay of HRI with HSP90 and limits the recruitment of HRI and HSP90 to unique p62/SQSTM1 complexes that also contain critical kinases like mTOR complex 1 and TBK1, and proteins involved in RNA metabolism, oxidative damage, and proteostasis like TDP-43. Thus, ROCK1 helps B cells cope with intense pathogen-driven destruction by coordinating the activity of key controllers of B cell differentiation and stress responses. These ROCK1-dependent mechanisms may be widely employed by cells to handle severe environmental stresses, and these findings may be relevant for immune-mediated and age-related neurodegenerative disorders.
Collapse
Affiliation(s)
- Juan Rivera-Correa
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, New York, USA
- Biological Sciences Department, New York City College of Technology, City University of New York, Brooklyn, New York, USA
| | - Sanjay Gupta
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, New York, USA
| | - Edd Ricker
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, New York, USA
| | - Danny Flores-Castro
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, New York, USA
| | - Daniel Jenkins
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, New York, USA
| | - Stephen Vulcano
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, New York, USA
| | - Swati P. Phalke
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, New York, USA
| | - Tania Pannellini
- Research Division and Precision Medicine Laboratory, Hospital for Special Surgery, New York, New York, USA
| | - Matthew M. Miele
- Microchemistry & Proteomics Core at Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Zhuoning Li
- Microchemistry & Proteomics Core at Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Nahuel Zamponi
- Hematology and Oncology Division, Weill Cornell Medicine, New York, New York, USA
| | - Young-Bum Kim
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Yurii Chinenov
- David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
| | - Eugenia Giannopoulou
- Biological Sciences Department, New York City College of Technology, City University of New York, Brooklyn, New York, USA
- David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
| | - Leandro Cerchietti
- Hematology and Oncology Division, Weill Cornell Medicine, New York, New York, USA
| | - Alessandra B. Pernis
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, New York, USA
- David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
- Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|