1
|
Xu NN, Wen YC, Pan J, Shu F, Qu JX, Qi XF, Tang J. Activated mTORC1 signaling pathway aggravates cisplatin induced oxidative damage by inhibiting autophagy in mouse cochlear hair cells. Neuropharmacology 2025; 272:110433. [PMID: 40147638 DOI: 10.1016/j.neuropharm.2025.110433] [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: 11/22/2024] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
Platinum-based antitumor drugs, such as cisplatin and carboplatin, are well-known for their severe ototoxicity. The ototoxic effects of these drugs are primarily attributed to oxidative stress induced damage within cochlear hair cells (HCs), leading to cell death and subsequent irreversible hearing loss. Over the past decade, studies have demonstrated that upregulating autophagy levels in HCs can greatly alleviate the death of cochlear HCs as part of the oxidative damage induced by ototoxic drugs. However, the molecular mechanisms by which platinum-based drugs affect autophagy and ultimately lead to HCs death remain unclear. In the present study, we investigated the effects of cisplatin on the mTOR signaling pathway, a critical regulator of autophagy, in cochlear explants of mice. Our results indicated that while cisplatin enhances autophagy activity initially, it also activates mTOR Complex1 (mTORC1) within HCs. The persistent activation of mTORC1 inhibits autophagy in HCs, resulting in the accumulation of reactive oxygen species and leading to cell death. Further pharmacological experiments confirmed the protective role of rapamycin, a specific mTORC1 inhibitor, highlighting the importance of autophagy in combating cisplatin-induced ototoxicity. Our findings suggest that modulating the mTOR signaling pathway to regulate autophagy could be an effective strategy for preventing cisplatin-induced ototoxic damage.
Collapse
Affiliation(s)
- Na-Na Xu
- Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yin-Chuan Wen
- Department of Anesthesiology, Shenzhen Maternity & Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, 518000, China
| | - Jing Pan
- Department of Otolaryngology Head & Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China; Ear Research Institute, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Fan Shu
- Department of Otolaryngology Head & Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China; Ear Research Institute, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Jia-Xi Qu
- Department of Otolaryngology Head & Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China; Ear Research Institute, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Xiao-Fei Qi
- Department of Anesthesiology, Shenzhen Maternity & Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, 518000, China.
| | - Jie Tang
- Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China; Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, 510515, China; Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
2
|
Ashtary-Larky D. Are plant-based and omnivorous diets the same for muscle hypertrophy? A narrative review of possible challenges of plant-based diets in resistance-trained athletes. Nutrition 2025; 135:112742. [PMID: 40215782 DOI: 10.1016/j.nut.2025.112742] [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: 11/21/2024] [Revised: 02/19/2025] [Accepted: 03/05/2025] [Indexed: 05/18/2025]
Abstract
This narrative review examines the potential challenges associated with plant-based diets in supporting muscle hypertrophy among resistance-trained athletes. Contrary to common assumptions, current evidence suggests that plant-based diets, when properly planned, can provide protein comparable to omnivorous diets. However, plant-based proteins are generally considered less anabolic due to lower digestibility, essential amino acid (EAA) content, and particularly lower leucine levels. The review discusses challenges and solutions for athletes aiming to maximize hypertrophy through plant-based diets, while highlighting the need for more robust research on advanced resistance-trained athletes.
Collapse
Affiliation(s)
- Damoon Ashtary-Larky
- Nutrition and Metabolic Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
3
|
Xu S, Zhang Z, Zhou X, Liao Y, Peng Z, Meng Z, Nüssler AK, Ma L, Xia H, Liu L, Yang W. Gouqi-derived Nanovesicles (GqDNVs) promoted MC3T3-E1 cells proliferation and improve fracture healing. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156755. [PMID: 40252435 DOI: 10.1016/j.phymed.2025.156755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 03/25/2025] [Accepted: 04/10/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Lycium barbarum L., also known as Gouqi, a traditional Chinese herbal medicine, is widely utilized in health care products and clinical therapies. Its muscle and bone strengthening efficacy has been recorded in medical classics for a long time. In addition, plant exosome-like nanovesicles (PELNVs) have attracted more and more attention owing to their biological traits. Therefore, we intended to explore the functions, regulatory role, and underlying mechanism of Gouqi-derived Nanovesicles (GqDNVs) on fracture healing. METHODS In this study, we employed the sucrose density gradient differential ultracentrifugation to isolate GqDNVs. The effects of GqDNVs on the proliferation and differentiation of MC3T3-E1 cells were evaluated using the CCK-8 assay, ALP activity measurement, and cell scratch assay. Additionally, leveraging a fracture mouse model, we utilized Micro-CT, immunological staining, and histologic analyses to comprehensively assess the impact of GqDNVs on fracture healing in mice. RESULTS GqDNVs stimulated cell viability, increased ALP activity, and promoted cellular osteogenic protein expression (OPN, ALP, and RUNX2). Subsequently, in the mouse fracture model, trabecular thickness, and bone marrow density were increased in the GqDNVs treatment group after 28 days of injection. Meanwhile, the expressions of OPN and BGP were significantly elevated after both 14 and 28 days. Additionally, the expressions of p-PI3K/PI3K, p-Akt/Akt, p-mTOR/mTOR, p-4EBP1/4EBP1 and p-p70S6K/ p70S6K were also increased after14 days of treatment. CONCLUSIONS GqDNVs effectively promoted the proliferation and differentiation of MC3T3-E1 cells. Furthermore, GqDNVs could improve fracture healing, which is associated with PI3K/Akt/mTOR/p70S6K/4EBP1 signaling pathway.
Collapse
Affiliation(s)
- Shiyin Xu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Zixuan Zhang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Xiaolei Zhou
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Yuxiao Liao
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Zhao Peng
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Zitong Meng
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Andreas K Nüssler
- Department of Traumatology, BG Trauma Center, University of Tübingen, Schnarrenbergstr. 95, Tübingen 72076, Germany
| | - Liang Ma
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hui Xia
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, China
| | - Liegang Liu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Wei Yang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China.
| |
Collapse
|
4
|
Liao HJ, Hong JR. Enhancement of reactive oxygen species metabolism by antioxidant enzyme Cu/Zn-SOD can block betanodavirus-induced necroptosis and suppress viral replication in fish cells. FISH & SHELLFISH IMMUNOLOGY 2025; 162:110344. [PMID: 40254084 DOI: 10.1016/j.fsi.2025.110344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/22/2025] [Accepted: 04/14/2025] [Indexed: 04/22/2025]
Abstract
Very little is known about oxidative stress as a modulator of signaling between the host and virus in viral nervous necrosis (VNN) within the aquaculture field. In the present study, we examined whether blocking ROS signaling using mitochondrial Cu/Zn-SOD could affect host cell death and the viral replication of RGNNV during infection in fish cells. Upon the overexpression of Cu/Zn-SOD in fish GF-1 cells, superoxide generation in RGNNV infection was reduced 0.6-fold, which correlated to host cell viability in the middle-late stages. Regarding the regulation of reactive oxygen species (ROS) signaling by superoxide, Cu/Zn-SOD overexpression can enhance superoxide's metabolism to hydrogen peroxide, which suppresses the RIPK3-mediated cell death signals at 48 hpf. On the other hand, ROS-mediated signal suppression can enhance Bcl-2 family Bcl-2/Bcl-xL expression in the early and middle replication stages. Furthermore, the enhancement of superoxide metabolism can reduce the virus' replication ability and expression of the non-structural genes B1 and B2, as well as the genome replication gene Protein A and the major capsid protein protein α, which were correlated with the viral load dropping by two log viral titers at 48 and 72 hpf. Taken together, these data suggest that ROS signals trigger host stress responses related to cell death/necroptosis in RGNNV infection. Then, ROS-mediated stress signals can modulate anti-cell death signals through the Bcl-2/Bcl-xL pathway. In conclusion, an ROS-mediated stress response is required for viral expression and replication cycles, providing new insights into controlling RNA viruses.
Collapse
Affiliation(s)
- Hong-Jun Liao
- Laboratory of Molecular Virology and Biotechnology, Institute of Biotechnology, National Cheng Kung University, Tainan, 701, Taiwan, ROC
| | - Jiann-Ruey Hong
- Laboratory of Molecular Virology and Biotechnology, Institute of Biotechnology, National Cheng Kung University, Tainan, 701, Taiwan, ROC; Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, 701, Taiwan, ROC.
| |
Collapse
|
5
|
Takano J, Takemoto D, Tatebe H, Shoji S, Fukuda K, Kitagawa Y, Rogi T, Izumo T, Nakao Y, Ishido M, Yoshimori T. Monocatechol metabolites of sesamin and episesamin promote higher autophagy flux compared to their unmetabolized forms by mTORC1-selective inhibition. Biochem Biophys Res Commun 2025; 765:151816. [PMID: 40279799 DOI: 10.1016/j.bbrc.2025.151816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/04/2025] [Accepted: 04/13/2025] [Indexed: 04/29/2025]
Abstract
Sesamin and episesamin, the major lignans found in refined sesame oil, reportedly exert antioxidant, anti-inflammatory, and hypocholesterolemic effects. Sesamin has also been suggested by previous studies to promote autophagy; however, concerns have been raised regarding the use of non-physiological concentrations, inaccurate methods for evaluating autophagic activity, and incomplete understanding of underlying mechanisms. Additionally, the effects of its metabolic kinetics on autophagy remain unclear. In this study, we demonstrated that sesamin, episesamin, and their metabolites induced autophagy flux at physiological concentrations in human cell cultures expressing monomeric red fluorescent protein-green fluorescent protein tandem fluorescent-tagged microtubule-associated protein 1A/1B-light-chain 3 proteins, a robust method for monitoring autophagy flux. Notably, the monocatechol metabolites of sesamin and episesamin exhibited higher autophagy flux than their unmetabolized forms. Immunoblotting analysis revealed that sesamin and its monocatechol metabolite promoted autophagy by inhibiting mammalian target of rapamycin complex 1 (mTORC1), leading to decreased phosphorylation of unc-51 like autophagy activating kinase 1 and transcription factor EB. This suppression enhanced the isolation membrane formation and transcriptionally stimulated autophagy and lysosomal biogenesis. Importantly, mTORC1 inhibition by sesamin and its metabolites did not affect mTORC2 activity, mirroring the mTORC1-selective inhibition observed with rapamycin. These results suggest that sesamin and episesamin contribute to diverse biological activities via their metabolism in the human body, regulating autophagy and mTORC1 signaling pathways.
Collapse
Affiliation(s)
- Jiro Takano
- Institute for Science of Life, Suntory Wellness Limited, Kyoto, 619-0284, Japan
| | - Daisuke Takemoto
- Institute for Science of Life, Suntory Wellness Limited, Kyoto, 619-0284, Japan.
| | | | | | | | - Yoshinori Kitagawa
- Institute for Science of Life, Suntory Wellness Limited, Kyoto, 619-0284, Japan
| | - Tomohiro Rogi
- Institute for Science of Life, Suntory Wellness Limited, Kyoto, 619-0284, Japan
| | - Takayuki Izumo
- Institute for Science of Life, Suntory Wellness Limited, Kyoto, 619-0284, Japan
| | - Yoshihiro Nakao
- Institute for Science of Life, Suntory Wellness Limited, Kyoto, 619-0284, Japan
| | | | - Tamotsu Yoshimori
- Health Promotion System Science, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
6
|
Aliyari M, Ghoflchi S, Hashemy SI, Hashemi SF, Reihani A, Hosseini H. The PI3K/Akt pathway: a target for curcumin's therapeutic effects. J Diabetes Metab Disord 2025; 24:52. [PMID: 39845908 PMCID: PMC11748622 DOI: 10.1007/s40200-025-01563-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 01/05/2025] [Indexed: 01/24/2025]
Abstract
Purpose The purpose of this review study is to investigate the effect of curcumin on the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) signaling pathway in various diseases. Curcumin, the main compound found in turmeric, has attracted a lot of attention for its diverse pharmacological properties. These properties have increased the therapeutic potential of curcumin in chronic diseases such as cardiovascular disease, Type 2 diabetes, obesity, non-alcoholic fatty liver disease, kidney disease, and neurodegenerative diseases. One of the main mechanisms of the effect of curcumin on health is its ability to modulate the PI3K/Akt signaling pathway. This pathway plays an important role in regulating vital cellular processes such as growth, cell survival, metabolism, and apoptosis. Disruption of the PI3K/Akt signaling pathway is associated with the incidence of several diseases. Methods Electronic databases including PubMed, Google Scholar, and Scopus were searched with the keywords "phosphoinositide 3-kinase" AND "protein kinase B "AND "curcumin" in the title/abstract. Also, following keywords "non-alcoholic fatty liver disease" AND "diabetes" AND "obesity" AND "kidney disease" and "neurodegenerative diseases" was searched in the whole text. Results Research indicates that curcumin offers potential benefits for several health conditions. Studies have shown it can help regulate blood sugar, reduce inflammation, and protect the heart, kidneys, and brain. Conclusion This protective effect is partially achieved by regulating the PI3K-Akt survival pathway, which helps improve metabolic disorders and oxidative stress. By examining how curcumin affects this vital cell pathway, researchers can discover new treatment strategies for a range of diseases.
Collapse
Affiliation(s)
- Mahdieh Aliyari
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sahar Ghoflchi
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Isaac Hashemy
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyedeh Fatemeh Hashemi
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirali Reihani
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Hosseini
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
7
|
Wang Y, Wu H, Hu X. Quantification of the inputs and outputs of serine and glycine metabolism in cancer cells. Arch Biochem Biophys 2025; 768:110367. [PMID: 40032043 DOI: 10.1016/j.abb.2025.110367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/10/2024] [Accepted: 02/28/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND The significance of serine and glycine metabolism in cancer cells is increasingly acknowledged, yet the quantification of their metabolic flux remains incomplete, impeding a comprehensive understanding. This study aimed to quantify the metabolic flux of serine and glycine in cancer cells, focusing on their inputs and outputs, by means of Combinations of C-13 Isotopes Tracing and mathematical delineation, alongside Isotopically Nonstationary Metabolic Flux Analysis. RESULTS In HeLa cells, serine uptake, the serine synthesis pathway (SSP), and other sources (e.g., protein degradation) contribute 71.2 %, 24.0 %, and 5.7 %, respectively, to serine inputs. Conversely, glycine inputs stem from uptake (45.6 %), conversion from serine (45.1 %), and other sources (9.4 %). Serine input flux surpasses glycine by 7.3-fold. Serine predominantly directs a major fraction (94.7 %) to phospholipid, sphingolipid, and protein synthesis, with only a minor fraction (5.3 %) directing towards one-carbon unit and glycine production. Glycine mainly supports protein and nucleotide synthesis (100 %), without conversion back to serine. Serine output rate exceeds glycine output rate by 7.3-fold. Serine deprivation mainly impairs output to synthesis of phospholipid and sphingolipid, crucial for cell growth, while other outputs unaffected. AGS cells exhibit comparable serine and glycine flux to HeLa cells, albeit lacking SSP activity. Serine deprivation in AGS cells halts output flux to phospholipid, sphingolipid, protein synthesis, completely inhibiting cell growth. CONCLUSIONS By providing quantitative insights into serine and glycine metabolism, this study delineates the association of serine flux to different metabolic pathway with cancer cell growth and offers potential targets for therapeutic intervention, highlighting the importance of serine flux to pathway for the synthesis of phospholipids and sphingolipids in cancer cells growth.
Collapse
Affiliation(s)
- Yuqi Wang
- Cancer Institute (Key Laboratory for Cancer Intervention and Prevention, China National Ministry of Education, Zhejiang Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, China; Department of Gastroenterology, The Second Affiliated Hospital, Zhejiang University School of Medicine, China
| | - Hao Wu
- Cancer Institute (Key Laboratory for Cancer Intervention and Prevention, China National Ministry of Education, Zhejiang Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, China.
| | - Xun Hu
- Cancer Institute (Key Laboratory for Cancer Intervention and Prevention, China National Ministry of Education, Zhejiang Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, China.
| |
Collapse
|
8
|
Liu J, Cui S, Ye Z, Chen J, Tang M, Chen C, Xu Y, Wang Z, Yang W, Zhang Z, Wang X. Transcriptomic analysis reveals the hepatopancreas metabolic mechanisms of mud crab Scylla paramamosain fed diets with terrestrial animal fat sources replacing fish oil. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2025; 54:101435. [PMID: 39922112 DOI: 10.1016/j.cbd.2025.101435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 02/03/2025] [Accepted: 02/03/2025] [Indexed: 02/10/2025]
Abstract
The transcriptome analysis following an 8-week feeding trial was employed to investigate the impacts of dietary terrestrial animal fats (TAFs includes lard oil (LO), beef tallow (BT) and poultry oil (PO)) replacing fish oil (FO) on the metabolic mechanism in hepatopancreas of mud crabs (Scylla paramamosain). The fatty acid (FA) transport, biosynthesis and lipid absorption and digestion were reduced through the regulation of PPAR pathway and the mRNA expressions of monoglyceride lipases (mgls), phosphatidate phosphatase-1 (pap1), acyl-sn-glycerol-3-phosphate acyltransferase delta (plcd), cAMP-dependent protein kinase catalytic (pkac), FA-binding protein 1 (fabp-1), FA transport protein 4 (fatp-4), short/branched chain specific acyl-CoA dehydrogenase (acdsb) and enoyl-CoA delta isomerase 2 (eci2), etc., after replacing FO with BT or LO. At the same time, dietary BT and LO regulated glycolysis, gluconeogenesis and insulin signals through increasing the genes of pyruvate dehydrogenase E1 (pdh), phosphoenolpyruvate carboxykinase (pepck) and phosphatidylinositol 3-kinase (pi3k) and regulated immunity status by down regulating the mRNA expressions of heat shock proteins 27 (hsp 27), cytochrome P450 (cyp 450), etc. Replacing FO with PO enhanced phospholipid storage, fat deposition, and inhibited glucose transport by up regulating pap1, mgls, lipin 1, lipinβ and down regulating glycosyl transferase (gt) and glucose transporter type 4 (glut4) expressions. The present study showed the signaling pathways and genes that were significantly regulated by TAFs replacing dietary FO, and revealed molecular mechanisms of TAFs in S. paramamosain. This would be conducive to the application of TAFs in aquatic feed.
Collapse
Affiliation(s)
- Jinjin Liu
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Shihui Cui
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Zihao Ye
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Jing Chen
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Mengyao Tang
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Chaojia Chen
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Yifang Xu
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Ziyi Wang
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Wei Yang
- Fujian Key Laboratory of Functional Aquafeed and Culture Environment Control, China
| | - Ziping Zhang
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| | - Xuexi Wang
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Fujian Key Laboratory of Functional Aquafeed and Culture Environment Control, China.
| |
Collapse
|
9
|
Gorjão N, Borowski LS, Szczesny RJ, Graczyk D. POLR1D, a shared subunit of RNA polymerase I and III, modulates mTORC1 activity. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119957. [PMID: 40222657 DOI: 10.1016/j.bbamcr.2025.119957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 03/21/2025] [Accepted: 04/08/2025] [Indexed: 04/15/2025]
Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) is a crucial nutrient sensor and a major regulator of cell growth and proliferation. While mTORC1 activity is frequently upregulated in cancer, the mechanisms regulating mTORC1 are not fully understood. POLR1D, a shared subunit of RNA polymerases I and III, is often upregulated in colorectal cancer (CRC) and mutated in Treacher-Collins syndrome. POLR1D, together with its binding partner POLR1C, forms a dimer that is believed to initiate the assembly of the multisubunit RNA polymerases I and III. Our data reveal an unexpected link between POLR1D and mTORC1 signalling. We found that the overproduction of POLR1D in human cells stimulates mTORC1 activity. In contrast, the downregulation of POLR1D leads to the repression of the mTORC1 pathway. Additionally, we demonstrate that a pool of POLR1D localises to the cytoplasm and interacts with the mTORC1 regulator RAGA and RAPTOR. Furthermore, POLR1D enhances the interaction between RAPTOR and RAGA and sustains mTORC1 activity under starvation conditions. We have identified a novel role for the RNA polymerase I/III subunit POLR1D in regulating mTORC1 signalling. Our findings suggest the existence of a new node in the already complex mTORC1 signalling network, where POLR1D functions to convey the cell's internal status, namely polymerase assembly, to this kinase.
Collapse
Affiliation(s)
- Neuton Gorjão
- Institute of Biochemistry and Biophysics Polish Academy of Sciences, ul. Pawińskiego 5a, 02-106 Warsaw, Poland
| | - Lukasz S Borowski
- Institute of Biochemistry and Biophysics Polish Academy of Sciences, ul. Pawińskiego 5a, 02-106 Warsaw, Poland; University of Warsaw, Faculty of Biology, Institute of Genetics and Biotechnology, ul. Pawińskiego 5a, 02-106 Warsaw, Poland
| | - Roman J Szczesny
- Institute of Biochemistry and Biophysics Polish Academy of Sciences, ul. Pawińskiego 5a, 02-106 Warsaw, Poland
| | - Damian Graczyk
- Institute of Biochemistry and Biophysics Polish Academy of Sciences, ul. Pawińskiego 5a, 02-106 Warsaw, Poland.
| |
Collapse
|
10
|
Yamada Y, Urakawa N, Tamiya H, Sakamoto S, Takahashi H, Harada N, Kitakaze T, Izawa T, Matsumua S, Yoshihara E, Inui H, Mashimo T, Yamaji R. Nrf2- and p53-inducible REDD2/DDiT4L/Rtp801L confers pancreatic β-cell dysfunction, leading to glucose intolerance in high-fat diet-fed mice. J Biol Chem 2025:110271. [PMID: 40409543 DOI: 10.1016/j.jbc.2025.110271] [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: 01/10/2025] [Revised: 05/07/2025] [Accepted: 05/11/2025] [Indexed: 05/25/2025] Open
Abstract
Pancreatic β-cells play a critical role in glucose homeostasis by secreting insulin. Chronic oxidative stress causes β-cell dysfunction, including β-cell loss; however, the underlying mechanisms remain unclear. Here, we demonstrate the critical role of the regulated in development and DNA damage response 2 (REDD2/DDiT4L/Rtp801L) in β-cell dysfunction. In INS-1 β-cells, Redd2 was induced by high glucose/palmitate or streptozotocin (STZ) exposure. Knockdown of Redd2 attenuated STZ-induced loss of cell viability, while REDD2 overexpression reduced cell viability and p70S6K phosphorylation, suggesting the involvement of suppression of mTORC1 activation. STZ also activated the transcription factors nuclear factor erythroid 2-related factor 2 (Nrf2) and p53, and overexpression of these transcription factors synergistically induced Redd2 expression. Reporter assays using the Redd2 promoter (-2328/-1) and chromatin immunoprecipitation identified the functional binding sites for Nrf2 (EpRE2, -349/-340) and p53 (p53RE1, -90/-81) on the Redd2 promoter. Purified recombinant p53 and Nrf2 bound directly. There were no noticeable changes in male global Redd2-knockout mice (C57BL/6J background), except for inguinal adipose tissue decrease when the mice were fed a standard diet. In contrast, when the mice were fed a high-fat diet (HFD), Redd2-knockout mice exhibited improved glucose tolerance relative to littermate controls. Redd2-knockout in HFD-fed mice increased β-cell mass due to reduced β-cell apoptosis and elevated plasma insulin concentrations, whereas insulin sensitivity remained unaffected. In both STZ-induced male and female and HFD-fed male models, β-cell specific Redd2-knockout improved glucose tolerance without affecting insulin sensitivity. Our results identify REDD2 as a novel regulator of β-cell dysfunction under oxidative stress.
Collapse
Affiliation(s)
- Yukiho Yamada
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Osaka Metropolitan University, Sakai, Osaka, Japan
| | - Natsuho Urakawa
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka, Japan
| | - Hisato Tamiya
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka, Japan
| | - Shuya Sakamoto
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka, Japan
| | - Hiroki Takahashi
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Osaka Metropolitan University, Sakai, Osaka, Japan
| | - Naoki Harada
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Osaka Metropolitan University, Sakai, Osaka, Japan; Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka, Japan; The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA.
| | - Tomoya Kitakaze
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Osaka Metropolitan University, Sakai, Osaka, Japan; Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka, Japan
| | - Takeshi Izawa
- Graduate School of Veterinary Science, Osaka Metropolitan University, Izumisano, Osaka, Japan
| | - Shigenobu Matsumua
- Department of Nutrition, Graduate School of Human Life and Ecology, Osaka Metropolitan University, Habikino, Osaka, Japan
| | - Eiji Yoshihara
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA; David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, USA
| | - Hiroshi Inui
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Osaka Metropolitan University, Sakai, Osaka, Japan; Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka, Japan; Department of Health and Nutrition, Otemae University, Osaka, Japan
| | - Tomoji Mashimo
- Division of Animal Genetics, Laboratory Animal Research Center, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo, Japan
| | - Ryoichi Yamaji
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Osaka Metropolitan University, Sakai, Osaka, Japan; Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka, Japan; Center for Research and Development of Bioresources, Osaka Metropolitan University, Sakai, Osaka, Japan
| |
Collapse
|
11
|
Wu J, Gao P, Yang C, Zhuang F, Luo Y, Wen F, Zhang P, Wang L, Xie H, Dai C, Zhao D, Li C, Deng H, Deng Z, Chen C. Targeting mitochondrial complex I of CD177 + neutrophils alleviates lung ischemia-reperfusion injury. Cell Rep Med 2025; 6:102140. [PMID: 40398393 DOI: 10.1016/j.xcrm.2025.102140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 01/01/2025] [Accepted: 04/24/2025] [Indexed: 05/23/2025]
Abstract
Primary graft dysfunction (PGD) is the leading cause of early morbidity and mortality following lung transplantation, with neutrophils playing a central role in its inflammatory pathology. Here, we employ single-cell RNA sequencing and spatial transcriptomics to investigate neutrophil subtypes in the lung ischemia-reperfusion injury (IRI) model. We identify CD177+ neutrophils as an activated subpopulation that significantly contributes to lung injury and serves as an early biomarker for predicting severe PGD in human lung transplant recipients (area under the curve [AUC] = 0.871). CD177+ neutrophils exhibit elevated oxidative phosphorylation and increased mitochondrial complex I activity, driving inflammation and the formation of neutrophil extracellular traps. Targeting mitochondrial function with the complex I inhibitor IACS-010759 reduces CD177+ neutrophil activation and alleviates lung injury in both mouse IRI and rat left lung transplant models. These findings provide a comprehensive landscape of CD177+ neutrophil-driven inflammation in lung IRI and highlight its potential value for future early diagnosis and therapeutic interventions.
Collapse
Affiliation(s)
- Junqi Wu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Peigen Gao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Chenlu Yang
- BGI Research, Beijing, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fenghui Zhuang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Yunzhe Luo
- BGI Research, Beijing, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | | | | | - Long Wang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Huikang Xie
- Department of Pathology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chenyang Dai
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Deping Zhao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Chongwu Li
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | | | | | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China.
| |
Collapse
|
12
|
Yuan W, Zhang H, Peng L, Chen C, Feng C, Tang Z, Cui P, Li Y, Li T, Qiu X, Cui Y, Zeng Y, Luo J, Xie X, Guo Y, Jiang X, Dai H. Inhibition of interferon regulatory factor 4 orchestrates T cell dysfunction, extending mouse cardiac allograft survival. Chin Med J (Engl) 2025; 138:1202-1212. [PMID: 38811343 PMCID: PMC12091588 DOI: 10.1097/cm9.0000000000003198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND T cell dysfunction, which includes exhaustion, anergy, and senescence, is a distinct T cell differentiation state that occurs after antigen exposure. Although T cell dysfunction has been a cornerstone of cancer immunotherapy, its potential in transplant research, while not yet as extensively explored, is attracting growing interest. Interferon regulatory factor 4 (IRF4) has been shown to play a pivotal role in inducing T cell dysfunction. METHODS A novel ultra-low-dose combination of Trametinib and Rapamycin, targeting IRF4 inhibition, was employed to investigate T cell proliferation, apoptosis, cytokine secretion, expression of T-cell dysfunction-associated molecules, effects of mitogen-activated protein kinase (MAPK) and mammalian target of rapamycin (mTOR) signaling pathways, and allograft survival in both in vitro and BALB/c to C57BL/6 mouse cardiac transplantation models. RESULTS In vitro , blockade of IRF4 in T cells effectively inhibited T cell proliferation, increased apoptosis, and significantly upregulated the expression of programmed cell death protein 1 (PD-1), Helios, CD160, and cytotoxic T lymphocyte-associated antigen (CTLA-4), markers of T cell dysfunction. Furthermore, it suppressed the secretion of pro-inflammatory cytokines interferon (IFN)-γ and interleukin (IL)-17. Combining ultra-low-dose Trametinib (0.1 mg·kg -1 ·day -1 ) and Rapamycin (0.1 mg·kg -1 ·day -1 ) demonstrably extended graft survival, with 4 out of 5 mice exceeding 100 days post-transplantation. Moreover, analysis of grafts at day 7 confirmed sustained IFN regulatory factor 4 (IRF4) inhibition, enhanced PD-1 expression, and suppressed IFN-γ secretion, reinforcing the in vivo efficacy of this IRF4-targeting approach. The combination of Trametinib and Rapamycin synergistically inhibited the MAPK and mTOR signaling network, leading to a more pronounced suppression of IRF4 expression. CONCLUSIONS Targeting IRF4, a key regulator of T cell dysfunction, presents a promising avenue for inducing transplant immune tolerance. In this study, we demonstrate that a novel ultra-low-dose combination of Trametinib and Rapamycin synergistically suppresses the MAPK and mTOR signaling network, leading to profound IRF4 inhibition, promoting allograft acceptance, and offering a potential new therapeutic strategy for improved transplant outcomes. However, further research is necessary to elucidate the underlying pharmacological mechanisms and facilitate translation to clinical practice.
Collapse
Affiliation(s)
- Wenjia Yuan
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Hedong Zhang
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Longkai Peng
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Chao Chen
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
- Medical College, Guangxi University, Nanning, Guangxi 530004, China
| | - Chen Feng
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Zhouqi Tang
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Pengcheng Cui
- Medical College, Guangxi University, Nanning, Guangxi 530004, China
| | - Yaguang Li
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Tengfang Li
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Xia Qiu
- Medical College, Guangxi University, Nanning, Guangxi 530004, China
| | - Yan Cui
- Medical College, Guangxi University, Nanning, Guangxi 530004, China
| | - Yinqi Zeng
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Jiadi Luo
- Department of Pathology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Xubiao Xie
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Yong Guo
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Xin Jiang
- Department of Organ Transplantation, The Fifth Clinical Medical College of Henan University of Chinese Medicine (Zhengzhou People’s Hospital), Zhengzhou, Henan 450000, China
| | - Helong Dai
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
- Medical College, Guangxi University, Nanning, Guangxi 530004, China
| |
Collapse
|
13
|
Wang X, Gao F, Guan J, Zhang L, Du L, Zhao Y, Gao F, Zhao K, He W, Lin J. mTOR blockade mitigates chemotherapy drug-induced intestinal toxicity via inhibition of pyroptosis. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167913. [PMID: 40398827 DOI: 10.1016/j.bbadis.2025.167913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 05/13/2025] [Accepted: 05/15/2025] [Indexed: 05/23/2025]
Abstract
Mammalian target of rapamycin (mTOR) signaling constitutes a crucial intracellular signaling pathway indispensable for regulating a variety of pathophysiological processes, including cancers. Intriguingly, the inhibition of mTOR can reverse the adverse effects induced by chemotherapy drugs; however, the fundamental mechanism underlying this phenomenon remains unclear. In this study, we demonstrate that mTOR signaling blockade can mitigate etoposide- or cisplatin-induced intestinal injury in mice. The mTOR inhibitor AZD8055 can inhibit chemotherapy drug-induced normal cell pyroptosis, as manifested by a decreased proportion of PI-positive cells, attenuated intestinal cell swelling, and reduced release of lactate dehydrogenase (LDH) and high mobility group box-1 protein (HMGB1). We further determined that mTOR inhibition suppressed the cleavage of caspase-3 and gasdermin E (GSDME), suggesting the inhibition of the caspase3/GSDME signaling pathway. We also discovered that AZD8055 can impede chemotherapy drug-induced alterations in mitochondrial membrane potential, reactive oxygen species generation, and DNA damage in intestinal cells, which are the key upstream events for activating caspase-3. Correspondingly, data from in vivo mouse models also demonstrated that AZD8055 effectively curtailed intestinal DNA damage and inflammation induced by chemotherapy drugs. Importantly, although AZD8055 counteracts the side effects of chemotherapy drugs, it does not substantially affect their anti-tumor activity. Our study proposes the potential application of mTOR inhibitors as chemoprotective agents, presenting a means to prolong the duration of chemotherapy drug use and optimize the chemotherapeutic regimen.
Collapse
Affiliation(s)
- Xinyue Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Fei Gao
- Department of Laboratory Animals, College of Animal Science, Jilin University, 130062 Changchun, China
| | - Jiyu Guan
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Lening Zhang
- Department of Thoracic Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin, China
| | - Li Du
- Department of Pulmonary and Critical Care Medicine, The Affiliate Hospital of Inner Mongolia Medical University, Hohhot 010050, Inner Mongolia Autonomous Region, China
| | - Yicheng Zhao
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China; Department of Thoracic Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin, China; Clinical Medical College, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Feng Gao
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Kui Zhao
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Wenqi He
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Jing Lin
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| |
Collapse
|
14
|
Rasmussen MB, Holgersen K, Muk T, Leto A, Stensballe A, van Hall G, Aunsholt L, Kappel SS, Zachariassen G, Sangild PT. Plasma amino acids after human milk fortification and associations with growth in preterm infants. Pediatr Res 2025:10.1038/s41390-025-04126-6. [PMID: 40383870 DOI: 10.1038/s41390-025-04126-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 04/14/2025] [Accepted: 04/18/2025] [Indexed: 05/20/2025]
Abstract
BACKGROUND It is unknown how plasma amino acid (AA) concentrations vary with fortification type, growth and insulin-like growth factor 1 (IGF-1) concentrations in the first weeks of life in very preterm infants (VPIs). METHODS Human milk for VPIs (n = 225) was fortified with bovine colostrum (BC, intact proteins, high bioactivity) or conventional fortifier (CF, hydrolysed bovine whey proteins). Plasma was sampled at fortification start (T0, ~1 week of age) and after one (T1) and two (T2) weeks. Changes in Z-scores for weight, length and head circumference (HC) were calculated from T0 to 35 weeks postmenstrual age. RESULTS Compared with CF, BC fortification increased 12 AAs (~10-40%, p < 0.05) and reduced Lys concentrations (10-16%, p < 0.05). Analysed across groups, T0-T2 AA increments associated positively with HC growth (12 AAs) and IGF-1 concentrations (5 AAs), and inversely with gestational age (13 AAs) and weight (8 AAs) at birth. The plasma protein profile (proteome) was unaffected by fortification. CONCLUSIONS BC fortification increased the plasma concentrations of many AAs. Fortification-induced AA increments associated positively with HC growth and IGF-1 concentrations, and were affected by immaturity and birth weight. Still, plasma AA variability within physiological levels appears to have limited implications for clinical outcomes during the early life of VPIs. IMPACT It is unknown how human milk fortification affects plasma amino acid concentrations, in turn influencing growth patterns in very preterm infants. We show that a fortifier based on bovine colostrum induces higher amino acid concentrations than a conventional fortifier. Fortification-induced increments in amino acid concentrations associated with gestational age, birth weight and head growth, but with small effect sizes and limited relation to body weight or length growth. Plasma amino acid concentrations are influenced by fortification of human milk in early life, but have limited practical application as predictors of body growth and health in individual very preterm infants.
Collapse
Affiliation(s)
- Martin Bo Rasmussen
- Comparative Pediatrics, Section for Biomedicine, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Kristine Holgersen
- Comparative Pediatrics, Section for Biomedicine, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
- Hans Christian Andersen Children's Hospital, Odense University Hospital, Odense, Denmark
| | - Tik Muk
- Comparative Pediatrics, Section for Biomedicine, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Azra Leto
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
- Sino-Danish Center, Beijing, China
| | - Allan Stensballe
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
- Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Gerrit van Hall
- Clinical Integrative Fluxomics core, Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lise Aunsholt
- Comparative Pediatrics, Section for Biomedicine, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
- Department of Neonatology, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Susanne Soendergaard Kappel
- Comparative Pediatrics, Section for Biomedicine, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
- Department of Neonatology, Rigshospitalet, Copenhagen, Denmark
| | - Gitte Zachariassen
- Hans Christian Andersen Children's Hospital, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Open Patient Explorative Network, Odense, Denmark
| | - Per Torp Sangild
- Comparative Pediatrics, Section for Biomedicine, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark.
- Hans Christian Andersen Children's Hospital, Odense University Hospital, Odense, Denmark.
- Department of Neonatology, Rigshospitalet, Copenhagen, Denmark.
- Centre for Science and Faith, Faculty of Theology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
15
|
Cheslock A, Provencher J, Campeau W, MacMillan HA. The impact of microplastics on tissue-specific gene expression in the tropical house cricket, Gryllodes sigillatus. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025:126475. [PMID: 40383478 DOI: 10.1016/j.envpol.2025.126475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 05/14/2025] [Accepted: 05/15/2025] [Indexed: 05/20/2025]
Abstract
Microplastics are ubiquitous in our environment, resulting in animal exposure and consumption via food, water, and air. Animals that consume microplastics may suffer from physiological effects like immunotoxicity or mitochondrial dysfunction, but how specific tissues may differentially respond to plastic consumption is poorly understood, particularly in terrestrial insects. Here, we measured transcriptomic responses of tissues (midgut, hindgut, fat body and ovaries) to microplastic consumption in a generalist ground-dwelling insect, the tropical house cricket, Gryllodes sigillatus. Using this approach, we provide insights on how microplastics may impact specific organ systems. We generated a de novo transcriptome, a useful resource for further studies on this emerging model insect, that we then used to infer differential gene expression due to microplastic consumption in individual organs. Ingestion of microplastics elicited unique changes in gene expression depending on the tissue of focus, with notable differentially-expressed genes related to survival and stress pathways as well as those related to metabolism, immunity, and cancer.
Collapse
Affiliation(s)
- Alexandra Cheslock
- Department of Biology, Carleton University, Ottawa, Ontario, Canada K1S 5B6
| | - Jennifer Provencher
- National Wildlife Research Centre, Environment Canada, Ottawa, Ontario, Canada
| | - Winston Campeau
- Department of Biology, Carleton University, Ottawa, Ontario, Canada K1S 5B6
| | - Heath A MacMillan
- National Wildlife Research Centre, Environment Canada, Ottawa, Ontario, Canada.
| |
Collapse
|
16
|
Dongye C, Chen X, Zhao Y, Li H, Abdallah MF, Li T, Chen X. Protective Effects of Octyl Gallate Against Deoxynivalenol-Induced Colon Inflammation: Insights from Proteomic and Metabolomic Analyses. ENVIRONMENT & HEALTH (WASHINGTON, D.C.) 2025; 3:515-525. [PMID: 40400552 PMCID: PMC12090014 DOI: 10.1021/envhealth.4c00250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 01/24/2025] [Accepted: 02/01/2025] [Indexed: 05/23/2025]
Abstract
Deoxynivalenol (DON) and octyl gallate (OG) are prevalent compounds in the environment and food. DON is frequently detected in cereals such as corn and wheat, while OG is commonly employed as a food additive. As a result, human exposure to these substances is inevitable. Given this, the objective of this experiment was to investigate the impact of co-exposure to DON (10 μg/kg) and OG (10 μg/kg) on intestinal inflammation. The RAW264.7 macrophage cell line was utilized to analyze cytokine levels as well as proteomic and metabolomic changes. In the quantitative real-time PCR experiments, the DON group showed significant difference compared to the control group (* p < 0.05) and the DON-OG group (# p < 0.05) regarding cytokine levels such as IL-10, TNF-α, Il6, Il1b, Ccl2, Il12α, Nos2, Cxcl1, and Cxcl2. In the animal experiment, C57BL/6 mice were utilized to monitor body weight, the presence of bloody stools, and diarrhea. Additionally, the colonic tissues of the mice underwent pathological analysis. The results indicated that cells treated with both DON and OG displayed lower levels of inflammation compared to those treated with DON alone. Furthermore, proteomic and metabolomic analyses revealed that the regulation of the Lancl2 protein and the mTOR signaling pathway contributed to the milder inflammatory response observed in the DON-OG group. These findings were further corroborated by the pathological analysis of the colonic tissues from the mice. In the combined exposure of DON and OG, OG partially mitigated the intestinal inflammation induced by DON.
Collapse
Affiliation(s)
- Chenxin Dongye
- Shandong
International Joint Laboratory of Environmental and Biological Science, Qilu University of Technology (Shandong Academy of
Science), Jinan, Shandong 250014, China
| | - Xiangrong Chen
- Shandong
International Joint Laboratory of Environmental and Biological Science, Qilu University of Technology (Shandong Academy of
Science), Jinan, Shandong 250014, China
| | - Yanfang Zhao
- Shandong
International Joint Laboratory of Environmental and Biological Science, Qilu University of Technology (Shandong Academy of
Science), Jinan, Shandong 250014, China
| | - Huijuan Li
- Shandong
International Joint Laboratory of Environmental and Biological Science, Qilu University of Technology (Shandong Academy of
Science), Jinan, Shandong 250014, China
| | - Mohamed F. Abdallah
- Department
of Human Biology and Toxicology, Faculty of Medicine, Pharmacy and
Biomedical Sciences, University of Mons, Mons 7000, Belgium
| | - Tianliang Li
- Center
for Cell Structure and Function, Shandong Provincial Key Laboratory
of Animal Resistance Biology, Collaborative Innovation Center of Cell
Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250358, China
| | - Xiangfeng Chen
- Shandong
International Joint Laboratory of Environmental and Biological Science, Qilu University of Technology (Shandong Academy of
Science), Jinan, Shandong 250014, China
| |
Collapse
|
17
|
Wang X, Zhong W, Wang Q, Song P, Lin X, Li B, Yin Y, Yang C, Li M. Lysionotin promoted apoptosis of hepatocellular carcinoma cells via inducing autophagy. Discov Oncol 2025; 16:788. [PMID: 40377756 PMCID: PMC12084452 DOI: 10.1007/s12672-025-02503-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 04/25/2025] [Indexed: 05/18/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma is a prevalent malignant tumor with a high mortality rate. Natural plants hold promise for its treatment, however, the mechanism of lysionotin induced apoptosis in liver cancer cells unclearly. This study aims to investigate the microenvironment alterations and the efficacy of lysionotin in liver cancer. METHODS Transmission electron microscopy, and laser confocal microscopy were employed to investigate the effect of lysionotin on autophagy in HCC cells. The molecular mechanism through which lysionotin induces autophagy and autophagy-induced apoptosis was ascertained by transcriptome sequencing, immunoblotting and Hoechst 33258 staining. RESULTS RNA sequencing analysis, electron microscopy and laser confocal microscopy revealed that lysionotin initiate autophagy in liver cancer cells. Immunoblotting indicated that lysionotin markedly enhances the activation of LC3-II in HCC cells, resulting in the activation of key effector molecules ATG12, Beclin-1 and the degradation of P62. Combined with autophagy inhibitors CQ and 3-MA significantly inhibited lysionotin-induced cell apoptosis. Immunoblotting and Hoechst staining disclosed that the activation of autophagy by lysionotin might be associated with the suppression of the mTOR-AKT signaling pathway. The treatment of mTOR inhibitor RAPA and activator 1485 demonstrated that inhibiting mTOR activation significantly augments the pro-apoptotic effect of lysionotin on liver cancer cells, while mTOR activator could rescue the effect of lysionotin on cells. CONCLUSIONS The findings suggest that the activation of autophagy by lysionotin may represent one of the pivotal mechanisms underlying its therapeutic efficacy against HCC and its synergistic enhancement of RAPA's antitumor effects.
Collapse
Affiliation(s)
- Xiaoxue Wang
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong, People's Republic of China
| | - Weiwei Zhong
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong, People's Republic of China
| | | | - Peng Song
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong, People's Republic of China
| | - Xia Lin
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong, People's Republic of China
| | - Bohan Li
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong, People's Republic of China
| | - Yancun Yin
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, Shandong, People's Republic of China
| | - Chunyan Yang
- School of Stomatology, Binzhou Medical University, Yantai, Shandong, People's Republic of China
| | - Minjing Li
- School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong, People's Republic of China.
| |
Collapse
|
18
|
Yu Y, Fu R, Jin C, Gao H, Han L, Qi M, Fu B, Li Q, Wang Y, Cheng Y, Leng J. Ruminal microbiome-host metabolome crosstalk in the synthesis of unsaturated fatty acids in buffalo milk. J Dairy Sci 2025:S0022-0302(25)00360-1. [PMID: 40383389 DOI: 10.3168/jds.2024-26176] [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: 01/04/2025] [Accepted: 04/24/2025] [Indexed: 05/20/2025]
Abstract
Interactions between rumen microorganisms and their metabolites contribute to milk yield and milk fat content in dairy cows. However, whether rumen microbes and host metabolism affect fatty acid synthesis in milk is unknown. In this study, we investigated the potential regulatory mechanisms affecting the unsaturated fatty acid content of Binglangjiang buffalo by using macrogenomics and metabolomics. Macrogenomic analysis showed that Bacteroides was significantly more abundant in the high UFA group (HF), contributing to the improvement of functions related to fatty acid synthesis. Then, we found that the rumen microbiota of the HF group was enriched in 2 important pathways involved in lipid metabolism (i.e., fatty acid biosynthesis and fatty acid metabolism), suggesting that more fatty acids were synthesized in the HF group. Metabolomics analyses showed that most of the UFA were more abundant in the HF group, which was also confirmed by the quantification of related metabolic pathways in milk fatty acids, suggesting that the HF group has a higher capacity to synthesize MUFA and PUFA. Correlation analysis of rumen lipid metabolic pathways and metabolites revealed that metabolic pathways such as fatty acid biosynthesis, fatty acid metabolism, metabolic pathways, and peroxisome proliferator-activated receptor (PPAR) signaling pathway, which were significantly enriched in the HF group compared with the low UFA group, were significantly and positively correlated with multiple UFA . The synthesis of UFA is mainly influenced by Bacteroides, Prevotella, and Bacteroidaceae, and regulated by fatty acid biosynthesis, fatty acid metabolism, and PPAR signaling pathways, which together influence the synthesis of UFA in buffaloes.
Collapse
Affiliation(s)
- Ye Yu
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; Key Laboratory of Animal Nutrition and Feed Science of Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
| | - Runqi Fu
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; Key Laboratory of Animal Nutrition and Feed Science of Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
| | - Chunjia Jin
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; Key Laboratory of Animal Nutrition and Feed Science of Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
| | - Huan Gao
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; Key Laboratory of Animal Nutrition and Feed Science of Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
| | - Lin Han
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; Key Laboratory of Animal Nutrition and Feed Science of Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
| | - Min Qi
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; Key Laboratory of Animal Nutrition and Feed Science of Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
| | - Binlong Fu
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; Key Laboratory of Animal Nutrition and Feed Science of Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
| | - Qian Li
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; Key Laboratory of Animal Nutrition and Feed Science of Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
| | - Yuyan Wang
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; Key Laboratory of Animal Nutrition and Feed Science of Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
| | - Yanfen Cheng
- Laboratory of Gastrointestinal Microbiology, National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Jing Leng
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; Key Laboratory of Animal Nutrition and Feed Science of Yunnan Province, Yunnan Agricultural University, Kunming 650201, China.
| |
Collapse
|
19
|
Zhang Y, Wu L, Zheng C, Xu H, Lin W, Chen Z, Cao L, Qu Y. Exploring potential diagnostic markers and therapeutic targets for type 2 diabetes mellitus with major depressive disorder through bioinformatics and in vivo experiments. Sci Rep 2025; 15:16834. [PMID: 40369032 PMCID: PMC12078483 DOI: 10.1038/s41598-025-01175-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 05/05/2025] [Indexed: 05/16/2025] Open
Abstract
Type 2 diabetes mellitus (T2DM) and Major depressive disorder (MDD) act as risk factors for each other, and the comorbidity of both significantly increases the all-cause mortality rate. Therefore, studying the diagnosis and treatment of diabetes with depression (DD) is of great significance. In this study, we progressively identified hub genes associated with T2DM and depression through WGCNA analysis, PPI networks, and machine learning, and constructed ROC and nomogram to assess their diagnostic efficacy. Additionally, we validated these genes using qRT-PCR in the hippocampus of DD model mice. The results indicate that UBTD1, ANKRD9, CNN2, AKT1, and CAPZA2 are shared hub genes associated with diabetes and depression, with ANKRD9, CNN2 and UBTD1 demonstrating favorable diagnostic predictive efficacy. In the DD model, UBTD1 (p > 0.05) and ANKRD9 (p < 0.01) were downregulated, while CNN2 (p < 0.001), AKT1 (p < 0.05), and CAPZA2 (p < 0.01) were upregulated. We have discussed their mechanisms of action in the pathogenesis and therapy of DD, suggesting their therapeutic potential, and propose that these genes may serve as prospective diagnostic candidates for DD. In conclusion, this work offers new insights for future research on DD.
Collapse
Affiliation(s)
- Yikai Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Linyue Wu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Chuanjie Zheng
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Huihui Xu
- Institute of Orthopedics and Traumatology, Zhejiang Provincial Hospital of Chinese Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Weiye Lin
- The First College of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zheng Chen
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Lingyong Cao
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| | - Yiqian Qu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| |
Collapse
|
20
|
Bozzi M, Sciandra F, Bigotti MG, Brancaccio A. Misregulation of the Ubiquitin-Proteasome System and Autophagy in Muscular Dystrophies Associated with the Dystrophin-Glycoprotein Complex. Cells 2025; 14:721. [PMID: 40422224 DOI: 10.3390/cells14100721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2025] [Revised: 05/09/2025] [Accepted: 05/12/2025] [Indexed: 05/28/2025] Open
Abstract
The stability of the sarcolemma is severely impaired in a series of genetic neuromuscular diseases defined as muscular dystrophies. These are characterized by the centralization of skeletal muscle syncytial nuclei, the replacement of muscle fibers with fibrotic tissue, the release of inflammatory cytokines, and the disruption of muscle protein homeostasis, ultimately leading to necrosis and loss of muscle functionality. A specific subgroup of muscular dystrophies is associated with genetic defects in components of the dystrophin-glycoprotein complex (DGC), which plays a crucial role in linking the cytosol to the skeletal muscle basement membrane. In these cases, dystrophin-associated proteins fail to correctly localize to the sarcolemma, resulting in dystrophy characterized by an uncontrolled increase in protein degradation, which can ultimately lead to cell death. In this review, we explore the role of intracellular degradative pathways-primarily the ubiquitin-proteasome and autophagy-lysosome systems-in the progression of DGC-linked muscular dystrophies. The DGC acts as a hub for numerous signaling pathways that regulate various cellular functions, including protein homeostasis. We examine whether the loss of structural stability within the DGC affects key signaling pathways that modulate protein recycling, with a particular emphasis on autophagy.
Collapse
Affiliation(s)
- Manuela Bozzi
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Sezione di Biochimica e Biochimica Clinica, Università Cattolica del Sacro Cuore, Largo F. Vito 1, 00168 Roma, Italy
- Istituto di Scienze e Tecnologie Chimiche "Giulio Natta"-SCITEC (CNR), Largo F. Vito, 00168 Roma, Italy
| | - Francesca Sciandra
- Istituto di Scienze e Tecnologie Chimiche "Giulio Natta"-SCITEC (CNR), Largo F. Vito, 00168 Roma, Italy
| | - Maria Giulia Bigotti
- Bristol Heart Institute, Bristol Royal Infirmary, Research Floor Level 7, Bristol BS2 8HW, UK
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - Andrea Brancaccio
- Istituto di Scienze e Tecnologie Chimiche "Giulio Natta"-SCITEC (CNR), Largo F. Vito, 00168 Roma, Italy
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| |
Collapse
|
21
|
Genc S, Cicek B. Examination of PDK1/AKT/mTOR transcription and exosomal mRNA levels in human glioblastoma cell line treated with a combination of temozolomide and hesperidin. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04137-4. [PMID: 40372477 DOI: 10.1007/s00210-025-04137-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 04/01/2025] [Indexed: 05/16/2025]
Abstract
The most malignant type of tumor in the brain is high-grade gliomas. Glioblastoma (GB), a grade 4 glioma, has the lowest 5-year survival rate and is associated with poor prognosis. An important signaling pathway involved in the pathogenesis of GB is the mammalian target of rapamycin (mTOR). Therefore, our study aimed to investigate how exosomes obtained from GB cells applied with different doses of hesperidin (HSP) affect miR- 9 and change the PDK1/AKT/mTOR pathway. For this purpose, T98G cells were treated with different doses (5, 10, 25, and 50 µg/mL) of HSP in combination with temozolomide (TMZ- 10 µg/mL). At the end of 24 h, cell viability, flow cytometry, and biochemical tests were performed. Additionally, exosomes were isolated from cells belonging to the control, TMZ, and high-concentration TMZ-HSP groups. miR- 9, PDK1, PTEN, AKT- 1, Bax, Bcl- 2, and Caspase 3 genes were expressed in both application groups and exosomes belonging to these groups. HSP was found to reduce the viability of GB cells significantly. The viability was significantly reduced, especially in the TMZ-HSP 50 µg/mL group. Depending on the dose, there was a significant increase in the LDH level and oxidative stress level. The apoptosis level was approximately 26% in the TMZ-HSP 50 µg/mL group. Along with all this, gene expressions changed at the exosomal level, and miR- 9 and miR- 146 levels increased. Similarly, it changed the expression of proteins related to the PDK1/AKT/mTOR signaling pathway at the exosomal level (p < 0.05). In conclusion, the TMZ-HSP combination showed anticancer effects in T98G cells, influenced exosome profiles, and appeared non-toxic and potentially beneficial to healthy cells, highlighting its potential therapeutic value.
Collapse
Affiliation(s)
- Sidika Genc
- Faculty of Medicine, Department of Medical Pharmacology, Bilecik Şeyh Edebali University, Bilecik, 11230, Turkey.
| | - Betul Cicek
- Faculty of Medicine, Department of Physiology, Erzincan Binali Yildirim University, Erzincan, 24100, Turkey
| |
Collapse
|
22
|
Ma JX, Li XJ, Li YL, Liu MC, Du RH, Cheng Y, Li LJ, Ai ZY, Jiang JT, Yan SY. Chaperonin-containing tailless complex polypeptide 1 subunit 6A negatively regulates autophagy and protects colorectal cancer cells from cisplatin-induced cytotoxicity. World J Gastroenterol 2025; 31:105729. [DOI: 10.3748/wjg.v31.i18.105729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/27/2025] [Accepted: 04/14/2025] [Indexed: 05/13/2025] Open
Abstract
BACKGROUND As a member of the chaperonin-containing tailless complex polypeptide 1 (TCP1) complex, which plays a pivotal role in ensuring the accurate folding of numerous proteins, chaperonin-containing TCP1 subunit 6A (CCT6A) participates in various physiological and pathological processes. However, its effects on cell death and cancer therapy and the underlying mechanisms need further exploration in colorectal cancer (CRC) cells.
AIM To explore the effects of CCT6A on cell death and cancer therapy and the underlying mechanisms in CRC.
METHODS Cell proliferation was evaluated using the MTS assay, EdU staining, and colony growth assays. The expression of CCT6A was monitored by immunoblotting and quantitative PCR. CCT6A was knocked out by CRISPR-Cas9, and overexpressed by transfecting plasmids. Autophagy was examined by immunoblotting and the mCherry-GFP-LC3 assay. To monitor apoptosis and necroptosis, immunoblotting, co-immunoprecipitation, and flow cytometry were employed.
RESULTS Cisplatin (DDP) exerted cytotoxic effects on CRC cells while simultaneously downregulating the expression of CCT6A. Depletion of CCT6A amplified the cytotoxic effects of DDP, whereas overexpression of CCT6A attenuated these adverse effects. CCT6A suppressed autophagy, apoptosis, and necroptosis under both basal and DDP-treated conditions. Autophagy inhibitors significantly enhanced the cytotoxic effects of DDP, whereas a necroptosis inhibitor partially reversed the cell viability loss induced by DDP. Furthermore, inhibiting autophagy enhanced both apoptosis and necroptosis induced by DDP.
CONCLUSION CCT6A negatively modulates autophagy, apoptosis, and necroptosis, and CCT6A confers resistance to DDP therapy in CRC, suggesting its potential as a therapeutic target.
Collapse
Affiliation(s)
- Jian-Xing Ma
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, Jining 272067, Shandong Province, China
| | - Xiao-Jun Li
- Department of General Surgery, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Ya-Long Li
- Department of General Surgery, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Ming-Chan Liu
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, Jining 272067, Shandong Province, China
| | - Rui-Hang Du
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, Jining 272067, Shandong Province, China
| | - Yi Cheng
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, Jining 272067, Shandong Province, China
| | - Liang-Jie Li
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, Jining 272067, Shandong Province, China
| | - Zhi-Ying Ai
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, Jining 272067, Shandong Province, China
| | - Jian-Tao Jiang
- The Second Affiliated Hospital of Xi’an Jiaotong University, Xibei Hospital, Xi’an 710000, Shaanxi Province, China
| | - Si-Yuan Yan
- Precision Medicine Laboratory for Chronic Non-communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, Jining 272067, Shandong Province, China
| |
Collapse
|
23
|
Delalat S, Sultana I, Osman H, Sieme M, Zhazykbayeva S, Herwig M, Budde H, Kovács Á, Kaçmaz M, Göztepe E, Borgmann N, Shahriari G, Sasko B, Wintrich J, Haldenwang P, Schmidt WE, Fenske W, Khan M, Jaquet K, Mügge A, Máthé D, Tóth VE, Varga ZV, Ferdinandy P, El-Battrawy I, van Heerebeek L, Hamdani N. Dysregulated inflammation, oxidative stress, and protein quality control in diabetic HFpEF: unraveling mechanisms and therapeutic targets. Cardiovasc Diabetol 2025; 24:211. [PMID: 40369521 PMCID: PMC12080046 DOI: 10.1186/s12933-025-02734-4] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Accepted: 04/07/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) represents a significant risk factor for cardiovascular disease, particularly heart failure with preserved ejection fraction (HFpEF). HFpEF predominantly affects elderly individuals and women, and is characterized by dysfunctions associated with metabolic, inflammatory, and oxidative stress pathways. Despite HFpEF being the most prevalent heart failure phenotype in patients with T2DM, its underlying pathophysiological mechanisms remain inadequately elucidated. OBJECTIVE This study aims to investigate the effects of diabetes mellitus on myocardial inflammation, oxidative stress, and protein quality control (PQC) mechanisms in HFpEF, with particular emphasis on insulin signaling, autophagy, and chaperone-mediated stress responses. METHODS We conducted an analysis of left ventricular myocardial tissue from HFpEF patients, both with and without diabetes, employing a range of molecular, biochemical, and functional assays. The passive stiffness of cardiomyocytes (Fpassive) was assessed in demembranated cardiomyocytes before and after implementing treatments aimed at reducing inflammation (IL-6 inhibition), oxidative stress (Mito-TEMPO), and enhancing PQC (HSP27, HSP70). Inflammatory markers (NF-κB, IL-6, TNF-α, ICAM-1, VCAM-1, NLRP3), oxidative stress markers (ROS, GSH/GSSG ratio, lipid peroxidation), and components of signaling pathways (PI3K/AKT/mTOR, AMPK, MAPK, and PKG) were evaluated using western blotting, immunofluorescence, and ELISA techniques. RESULTS Hearts from diabetic HFpEF patients exhibited significantly heightened inflammation, characterized by the upregulation of NF-κB, IL-6, and the NLRP3 inflammasome. This increase in inflammation was accompanied by elevated oxidative stress, diminished nitric oxide (NO) bioavailability, and impaired activation of the NO-sGC-cGMP-PKG signaling pathway. Notably, dysregulation of insulin signaling was observed, as indicated by decreased AKT phosphorylation and impaired autophagy regulation mediated by AMPK and mTOR. Additionally, PQC dysfunction was evidenced by reduced expression levels of HSP27 and HSP70, which correlated with increased cardiomyocyte passive stiffness. Targeted therapeutic interventions effectively reduced Fpassive, with IL-6 inhibition, Mito-TEMPO, and HSP administration leading to improvements in cardiomyocyte mechanical properties. CONCLUSION The findings of this study elucidate a mechanistic relationship among diabetes, inflammation, oxidative stress, and PQC impairment in the context of HFpEF. Therapeutic strategies that target these dysregulated pathways, including IL-6 inhibition, mitochondrial antioxidants, and chaperone-mediated protection, may enhance myocardial function in HFpEF patients with T2DM. Addressing these molecular dysfunctions could facilitate the development of novel interventions specifically tailored to the diabetic HFpEF population.
Collapse
Affiliation(s)
- Simin Delalat
- Medical Faculty, Department of Cellular and Translational Physiology, Institute of Physiology, Molecular and Experimental Cardiology, Institut Für Forschung und Lehre (IFL), Ruhr University Bochum, 44801, Bochum, Germany
| | - Innas Sultana
- Medical Faculty, Department of Cellular and Translational Physiology, Institute of Physiology, Molecular and Experimental Cardiology, Institut Für Forschung und Lehre (IFL), Ruhr University Bochum, 44801, Bochum, Germany
| | - Hersh Osman
- Medical Faculty, Department of Cellular and Translational Physiology, Institute of Physiology, Molecular and Experimental Cardiology, Institut Für Forschung und Lehre (IFL), Ruhr University Bochum, 44801, Bochum, Germany
| | - Marcel Sieme
- Medical Faculty, Department of Cellular and Translational Physiology, Institute of Physiology, Molecular and Experimental Cardiology, Institut Für Forschung und Lehre (IFL), Ruhr University Bochum, 44801, Bochum, Germany
| | - Saltanat Zhazykbayeva
- Medical Faculty, Department of Cellular and Translational Physiology, Institute of Physiology, Molecular and Experimental Cardiology, Institut Für Forschung und Lehre (IFL), Ruhr University Bochum, 44801, Bochum, Germany
| | - Melissa Herwig
- Medical Faculty, Department of Cellular and Translational Physiology, Institute of Physiology, Molecular and Experimental Cardiology, Institut Für Forschung und Lehre (IFL), Ruhr University Bochum, 44801, Bochum, Germany
| | - Heidi Budde
- Medical Faculty, Department of Cellular and Translational Physiology, Institute of Physiology, Molecular and Experimental Cardiology, Institut Für Forschung und Lehre (IFL), Ruhr University Bochum, 44801, Bochum, Germany
| | - Árpád Kovács
- Medical Faculty, Department of Cellular and Translational Physiology, Institute of Physiology, Molecular and Experimental Cardiology, Institut Für Forschung und Lehre (IFL), Ruhr University Bochum, 44801, Bochum, Germany
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Mustafa Kaçmaz
- Medical Faculty, Department of Cellular and Translational Physiology, Institute of Physiology, Molecular and Experimental Cardiology, Institut Für Forschung und Lehre (IFL), Ruhr University Bochum, 44801, Bochum, Germany
| | - Eda Göztepe
- Medical Faculty, Department of Cellular and Translational Physiology, Institute of Physiology, Molecular and Experimental Cardiology, Institut Für Forschung und Lehre (IFL), Ruhr University Bochum, 44801, Bochum, Germany
| | - Natalie Borgmann
- Medical Faculty, Department of Cellular and Translational Physiology, Institute of Physiology, Molecular and Experimental Cardiology, Institut Für Forschung und Lehre (IFL), Ruhr University Bochum, 44801, Bochum, Germany
| | - Gelareh Shahriari
- Medical Faculty, Department of Cellular and Translational Physiology, Institute of Physiology, Molecular and Experimental Cardiology, Institut Für Forschung und Lehre (IFL), Ruhr University Bochum, 44801, Bochum, Germany
| | - Benjamin Sasko
- Medical Department II, Marien Hospital Herne, Ruhr University Bochum, Bochum, Germany
| | - Jan Wintrich
- Medical Department II, Marien Hospital Herne, Ruhr University Bochum, Bochum, Germany
| | - Peter Haldenwang
- Department of Cardiothoracic Surgery, University Hospital Bergmannsheil Bochum, Bochum, Germany
| | - Wolfgang E Schmidt
- Department of Medicine I, St. Josef Hospital, UK RUB, Ruhr University Bochum, 44801, Bochum, Germany
| | - Wiebke Fenske
- Department of Internal Medicine I- General Internal Medicine, Endocrinology and Diabetology, Gastroenterology and Hepatology, BG University Hospital Bergmannsheil, Bochum, Germany
| | - Muchtiar Khan
- Department of Cardiology, OLVG, 1091 AC, Amsterdam, The Netherlands
| | - Kornelia Jaquet
- Medical Faculty, Department of Cellular and Translational Physiology, Institute of Physiology, Molecular and Experimental Cardiology, Institut Für Forschung und Lehre (IFL), Ruhr University Bochum, 44801, Bochum, Germany
| | - Andreas Mügge
- Medical Faculty, Department of Cellular and Translational Physiology, Institute of Physiology, Molecular and Experimental Cardiology, Institut Für Forschung und Lehre (IFL), Ruhr University Bochum, 44801, Bochum, Germany
| | - Domokos Máthé
- Department of Biophysics and Radiation Biology, Semmelweis University, Tűzoltó utca 37-47, 1094, Budapest, 1085, Hungary
- In Vivo Imaging Advanced Core Facility, Hungarian Centre of Excellence for Molecular Medicine, Budapest, Tűzoltó utca 37-47, 1094, Hungary
| | - Viktória E Tóth
- HCEMM-SU Cardiometabolic Immunology Research Group, Department of Pharmacology and pharmacotherapy,, Semmelweis University, Budapest, 1089, Hungary
- Center for Pharmacology and Drug Research & Development,Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, 1089, Hungary
| | - Zoltán V Varga
- HCEMM-SU Cardiometabolic Immunology Research Group, Department of Pharmacology and pharmacotherapy,, Semmelweis University, Budapest, 1089, Hungary
- Center for Pharmacology and Drug Research & Development,Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, 1089, Hungary
| | - Péter Ferdinandy
- HCEMM-SU Cardiometabolic Immunology Research Group, Department of Pharmacology and pharmacotherapy,, Semmelweis University, Budapest, 1089, Hungary
- Center for Pharmacology and Drug Research & Development,Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, 1089, Hungary
| | - Ibrahim El-Battrawy
- Medical Faculty, Department of Cellular and Translational Physiology, Institute of Physiology, Molecular and Experimental Cardiology, Institut Für Forschung und Lehre (IFL), Ruhr University Bochum, 44801, Bochum, Germany
- Department of Medicine I, St. Josef Hospital, UK RUB, Ruhr University Bochum, 44801, Bochum, Germany
| | | | - Nazha Hamdani
- Medical Faculty, Department of Cellular and Translational Physiology, Institute of Physiology, Molecular and Experimental Cardiology, Institut Für Forschung und Lehre (IFL), Ruhr University Bochum, 44801, Bochum, Germany.
- Department of Physiology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands.
- Medical Faculty, Department Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, MA 2/156, 44780, Bochum, Germany.
- Institut Für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Gudrunstraße 56, 44791, Bochum, Germany.
- Department of Cardiology, St. Josef-Hospital, UK RUB, Ruhr University Bochum, Bochum, Germany.
| |
Collapse
|
24
|
Joodi SA, Khattab MM, Ibrahim WW. Repurposing of cabergoline to improve cognitive decline in D-galactose-injected ovariectomized rats: Modulation of AKT/mTOR, GLT-1/P38-MAPK, and ERK1/2 signaling pathways. Toxicol Appl Pharmacol 2025; 500:117391. [PMID: 40349788 DOI: 10.1016/j.taap.2025.117391] [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: 01/25/2025] [Revised: 04/18/2025] [Accepted: 05/09/2025] [Indexed: 05/14/2025]
Abstract
Dopamine is involved in many physiological functions including reward phenomenon, motor, learning, and memory functions. Dopamine receptor agonists have been shown to reduce amyloid (Aβ) deposition, enhance memory, and improve cortical plasticity in experimental studies and Alzheimer's disease (AD) patients; however, the molecular mechanisms involved haven't been investigated yet. The target of this investigation was to elucidate the modulatory effects of cabergoline (CAB), a dopamine receptor agonist, against AD. Ovariectomized rats were injected with D-galactose (150 mg/kg/day, i.p) for ten weeks to exacerbate AD. CAB administration (1 mg/kg/day, i.p) for 28 days, beginning from the 7th week of D-galactose administration, attenuated the associated histopathological alterations and enhanced the spatial and recognition memory in Morris water maze and Novel object recognition tests, respectively. CAB decreased the hippocampal concentrations of Aβ42, p-tau, and β-secretase, while upregulating α-secretase. Moreover, CAB diminished nuclear factor-kappa β (NF-κβ), tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and myeloperoxidase, while elevating brain-derived neurotrophic factor and phospho-cAMP response element binding protein. Further, CAB reduced the hippocampal phosphorylated forms of protein kinase B (AKT) and mammalian target of rapamycin (mTOR) contrary to elevating Beclin-1, resulting in autophagy induction, which participates in accelerating Aβ42 and p-tau aggregates clearance. Moreover, CAB increased the hippocampal glutamate transporter-1 (GLT-1) protein expression, promoting glutamate uptake that possibly reduced Ca2+ overload and consequently decreased the phosphorylated forms of P38-MAPK and ERK1/2. In conclusion, CAB improved cognitive decline of D-gal/OVX animals, restored hippocampal architecture, exerted neuroprotection, and enhanced autophagic machinery via modulating AKT/mTOR, GLT-1/P38-MAPK, and ERK1/2 pathways.
Collapse
Affiliation(s)
- Sheer A Joodi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Mahmoud M Khattab
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Weam W Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| |
Collapse
|
25
|
Bertram JF, Cullen-McEwen LA, Andrade-Oliveira V, Câmara NOS. The intelligent podocyte: sensing and responding to a complex microenvironment. Nat Rev Nephrol 2025:10.1038/s41581-025-00965-y. [PMID: 40341763 DOI: 10.1038/s41581-025-00965-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2025] [Indexed: 05/11/2025]
Abstract
Podocytes are key components of the glomerular filtration barrier - a specialized structure that is responsible for the filtration of blood by the kidneys. They therefore exist in a unique microenvironment exposed to mechanical force and the myriad molecules that cross the filtration barrier. To survive and thrive, podocytes must continually sense and respond to their ever-changing microenvironment. Sensing is achieved by interactions with the surrounding extracellular matrix and neighbouring cells, through a variety of pathways, to sense changes in environmental factors such as nutrient levels including glucose and lipids, oxygen levels, pH and pressure. The response mechanisms similarly involve a range of processes, including signalling pathways and the actions of specific organelles that initiate and regulate appropriate responses, including alterations in cell metabolism, immune regulation and changes in podocyte structure and cognate functions. These functions ultimately affect glomerular and kidney health. Imbalances in these processes can lead to inflammation, podocyte loss and glomerular disease.
Collapse
Affiliation(s)
- John F Bertram
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia
- Australian Research Council Training Centre for Cell and Tissue Engineering Technologies, Brisbane, Queensland, Australia
| | - Luise A Cullen-McEwen
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia
| | - Vinicius Andrade-Oliveira
- Center for Natural and Human Sciences, Federal University of ABC, Sao Paulo, Brazil.
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil.
| | | |
Collapse
|
26
|
Wang L, Ma L, Gao Z, Wang Y, Qiu J. Significance of gene therapy in neurodegenerative diseases. Front Neurosci 2025; 19:1515255. [PMID: 40406043 PMCID: PMC12095248 DOI: 10.3389/fnins.2025.1515255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 04/10/2025] [Indexed: 05/26/2025] Open
Abstract
Gene therapy is an approach that employs vectors to deliver genetic material to target cells, aiming to correct genes with pathogenic mutations and modulate one or more genes responsible for disease progression. It holds significant value for clinical applications and offers broad market potential due to the large patient population affected by various conditions. For instance, in 2023, the Food and Drug Administration (FDA) approved 55 new drugs, including five specifically for gene therapy targeting hematologic and rare diseases. Recently, with advancements in understanding the pathogenesis and development of neurodegenerative diseases (NDDs), gene therapy has emerged as a promising avenue for treating Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), amyotrophic lateral sclerosis (ALS), and spinal muscular atrophy (SMA), particularly in personalized medicine. Notably, the FDA has approved three clinical applications for combating SMA, utilizing viral vectors delivered via intravenous and intrathecal injections. However, gene therapy for other NDDs remains in clinical trials, necessitating improvements in viral vectors, exploration of new vectors, optimization of delivery routes, and further investigation into pathogenesis to identify novel targets. This review discusses recent advancements in gene therapy for NDDs, offering insights into developing new therapeutic strategies.
Collapse
Affiliation(s)
- Lingling Wang
- Department of Neurology, Yantai Shan Hospital, Yantai, China
| | - Lin Ma
- Department of Neurology, Qingdao Municipal Hospital, Qingdao, China
| | - Zihan Gao
- Department of Internal Medicine of Traditional Chinese Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ying Wang
- Department of Neurology, Yantai Shan Hospital, Yantai, China
| | - Jiaoxue Qiu
- Department of Neurology, Yantai Shan Hospital, Yantai, China
| |
Collapse
|
27
|
Haidurov A, Budanov AV. Sestrins in Carcinogenesis-The Firefighters That Sometimes Stoke the Fire. Cancers (Basel) 2025; 17:1578. [PMID: 40361504 PMCID: PMC12071529 DOI: 10.3390/cancers17091578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2025] [Revised: 04/29/2025] [Accepted: 05/05/2025] [Indexed: 05/15/2025] Open
Abstract
Sestrins (SESN1-3) are a family of stress-responsive proteins that regulate cellular metabolism and redox balance, both of which are frequently disrupted in cancer. As direct targets of stress-responsive transcription factors, including tumour suppressor p53, Sestrins function as leucine-dependent inhibitors of mTORC1 and potent antioxidants. Their downregulation is widely observed across multiple cancers and is associated with increased tumour growth and poor prognosis. Despite their consistent tumour-suppressive effects through mTORC1 inhibition and promotion of p53-dependent apoptosis, Sestrins exhibit a limited role in tumour initiation, which appears to be context-dependent. Their antioxidant activity reduces oxidative damage, thereby protecting against genomic instability and other cancer-promoting events. However, in certain contexts, Sestrins may promote tumour survival and progression by stimulating pro-survival pathways, such as AKT signalling through mTORC2 activation. This review examines the molecular mechanisms underlying these dual functions, with a particular focus on mTOR signalling and oxidative stress. We also discuss Sestrin expression patterns and functional outcomes in various cancer types, including lung, liver, colon, skin, prostate, and follicular lymphomas, highlighting their potential as diagnostic markers and therapeutic targets.
Collapse
Affiliation(s)
- Alexander Haidurov
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Pearse Street, D02 R590 Dublin, Ireland
| | - Andrei V. Budanov
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Pearse Street, D02 R590 Dublin, Ireland
| |
Collapse
|
28
|
Abdel-Lah ES, Sherkawy HS, Mohamed WH, Fawy MA, Hasan AA, Muhammed AA, Taha AF, Tony AA, Hamad N, Gamea MG. Empagliflozin and memantine combination ameliorates cognitive impairment in scopolamine + heavy metal mixture-induced Alzheimer's disease in rats: role of AMPK/mTOR, BDNF, BACE-1, neuroinflammation, and oxidative stress. Inflammopharmacology 2025:10.1007/s10787-025-01755-5. [PMID: 40325262 DOI: 10.1007/s10787-025-01755-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Accepted: 04/02/2025] [Indexed: 05/07/2025]
Abstract
One of the major consequences of diabetes mellitus that has gained attention due to its rising incidence is cognitive impairment. Recent research suggested that sodium-glucose cotransporter-2 (SGLT-2) inhibitors can mitigate memory impairment linked to Alzheimer's disease and are now being explored for their cognitive benefits. However, their mechanisms were not thoroughly studied. This research investigates the hypothesis of the neuroprotective effect of empagliflozin administration against scopolamine-heavy metal mixture (SCO + HMM)-treated Alzheimer's rat models in comparison with memantine as a reference drug and the impact of their combination. Yet, the neuroprotective effects of memantine and empagliflozin combination against cognitive impairment have not been previously explored. This study employed adult male albino rats categorized into five groups. The impact of empagliflozin, memantine, and their concomitant administration on cognitive performance was assessed in a scopolamine and heavy metal mixture-treated Alzheimer's disease model in rats. The assessment of rats' cognitive behavior, memory, and spatial learning was conducted, followed by an evaluation of hippocampal brain-derived neurotrophic factor (BDNF), beta-secretase (BACE-1), oxidative stress (OS), and inflammatory marker activity. And, a western blot analysis was conducted to detect phosphorylated 5' AMP-activated protein kinase (p-AMPK), phosphorylated mammalian target of rapamycin (p-mTOR), and heme oxygenase-1 (HO-1). Hippocampal and cerebellar histopathology were thoroughly examined, in addition to the expressions of amyloid β (Aβ). The current data demonstrate the involvement of the pAMPK/mTOR/HO-1 signaling pathway in empagliflozin neuroprotection against SCO + HMM-induced AD. In addition, it reduces AD hallmarks (Aβ and BACE1), neuro-inflammation, and oxidative stress sequelae, and enhances neurogenesis and synaptic density via BDNF. This study proposes that EMPA, especially when co-administered with other conventional anti-Alzheimer therapy, may be formulated into an innovative therapeutic strategy for the enhancement of cognitive impairments associated with neurodegenerative disorders.
Collapse
Affiliation(s)
- Ebtsam S Abdel-Lah
- Department of Pharmacology, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt.
- Department of Pharmacology, School of Veterinary Medicine, Badr University, Assiut, 11829, Egypt.
| | - Hoda S Sherkawy
- Department of Medical Biochemistry, Faculty of Medicine, Aswan University, Aswan, Egypt
| | - Wafaa H Mohamed
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Aswan University, Aswan, Egypt
| | - Mariam A Fawy
- Department of Zoology, Faculty of Science, South Valley University, Qena, 83523, Egypt
| | - Asmaa A Hasan
- Department of Human Anatomy and Embryology, Faculty of Medicine, Aswan University, Aswan, 81528, Egypt
| | - Asmaa A Muhammed
- Department of Medical Physiology, Faculty of Medicine, Aswan University, Aswan, 81528, Egypt
| | - Amira F Taha
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Assiut University, Assiut, 71526, Egypt
| | - Abeer A Tony
- Department of Neuropsychiatry, Faculty of Medicine, Aswan University, Aswan, Egypt
| | - Nashwa Hamad
- Department of Pathology, Faculty of Veterinary Medicine, Assiut University, Assiut, 71515, Egypt
| | - Marwa G Gamea
- Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut, 71526, Egypt
- Basic Medical Science Department, Badr University, Assiut, Egypt
| |
Collapse
|
29
|
Xu Q, Wang J, Li Y, Lei H, Jin N, Lu J, Qian C, Zhang J, Dong J, Wang X. The inhibition of placental mTOR signaling leads to fetal growth restriction with abnormal glucose metabolism in different anatomical regions of placentas. Placenta 2025; 164:31-40. [PMID: 40090159 DOI: 10.1016/j.placenta.2025.03.008] [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: 09/22/2024] [Revised: 03/09/2025] [Accepted: 03/11/2025] [Indexed: 03/18/2025]
Abstract
INTRODUCTION Fetal growth restriction (FGR) is a common pregnancy complication with significant impact on obstetric and birth outcomes. Increasing evidence shows that the inhibition of placental mechanistic target of rapamycin (mTOR) signaling is closely related to FGR. However, the pathogenesis of FGR is not fully consistent presently, which is subject to the methodological divergence. METHODS Rapamycin was used to construct the FGR mouse model. Hematoxylin & eosin (HE) and periodic acid-schiff (PAS) staining were used to analyze the morphology of mouse placenta. Western blot was used to analyze the expression levels of glucose transporters and key enzymes associated with glycogen metabolism in human/mouse placental tissues in different anatomic layers. HTR-8 cells were treated with dimethyl sulfoxide (DMSO) or rapamycin (2 mM) for 24 h. Cell viability was detected by CCK8 kit. In addition, glycogen concentration in placental tissue or cell samples was detected by Glycogen Assay Kit. RESULTS Firstly, we observed a significant reduction of glucose content in different anatomical regions of human small-for-gestational-age (SGA) placenta, also glucose metabolism was undermined to some extent. Then, we found that FGR placentas showed abnormal morphological changes, the glycogen levels in FGR placentas were significantly reduced by quantitative detection. Meanwhile, the expression levels of glucose transporters, Gys1 and p-Gsk3β in FGR placentas were reduced compared to controls. The HTR-8 cells treated with rapamycin revealed decreasing mTOR activity and glycogen levels. In addition, glucose transporter, GYS1, p-GSK3β expressions were all significantly reduced and t-GSK3β level was significantly elevated. DISCUSSION Overall, our data indicate that inhibition of placental mTOR signaling may contribute to the occurrence of FGR by altering glucose metabolism.
Collapse
Affiliation(s)
- Qian Xu
- Department of Obstetrics and Gynecology, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi Province, China; Reproductive Medicine Department, General Hospital of Chinese PLA Central Theater Command, Wuhan, 430070, China
| | - Jingjing Wang
- Department of Obstetrics and Gynecology, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi Province, China
| | - Yajing Li
- Department of Obstetrics and Gynecology, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi Province, China
| | - Hui Lei
- Department of Obstetrics and Gynecology, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi Province, China
| | - Ni Jin
- Department of Obstetrics and Gynecology, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi Province, China
| | - Jie Lu
- Department of Obstetrics and Gynecology, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi Province, China
| | - Chenxi Qian
- Unit 95885 of the People's Liberation Army, China
| | - Jianhua Zhang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi Province, China
| | - Jie Dong
- Department of Obstetrics and Gynecology, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi Province, China.
| | - Xiaohong Wang
- Department of Obstetrics and Gynecology, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi Province, China.
| |
Collapse
|
30
|
Khalaf F, Barayan D, Saldanha S, Jeschke MG. Metabolaging: a new geroscience perspective linking aging pathologies and metabolic dysfunction. Metabolism 2025; 166:156158. [PMID: 39947519 DOI: 10.1016/j.metabol.2025.156158] [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: 11/06/2024] [Revised: 01/31/2025] [Accepted: 02/09/2025] [Indexed: 02/16/2025]
Abstract
With age, our metabolic systems undergo significant alterations, which can lead to a cascade of adverse effects that are implicated in both metabolic disorders, such as diabetes, and in the body's ability to respond to acute stress and trauma. To elucidate the metabolic imbalances arising from aging, we introduce the concept of "metabolaging." This framework encompasses the broad spectrum of metabolic disruptions associated with the hallmarks of aging, including the functional decline of key metabolically active organs, like the adipose tissue. By examining how these organs interact with essential nutrient-sensing pathways, "metabolaging" provides a more comprehensive view of the systemic metabolic imbalances that occur with age. This concept extends to understanding how age-related metabolic disturbances can influence the response to acute stressors, like burn injuries, highlighting the interplay between metabolic dysfunction and the ability to handle severe physiological challenges. Finally, we propose potential interventions that hold promise in mitigating the effects of metabolaging and its downstream consequences.
Collapse
Affiliation(s)
- Fadi Khalaf
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada; David Braley Research Institute, Hamilton, Ontario, Canada; Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Dalia Barayan
- David Braley Research Institute, Hamilton, Ontario, Canada; Hamilton Health Sciences, Hamilton, Ontario, Canada; Department of Surgery, McMaster University, Hamilton, Ontario, Canada
| | - Sean Saldanha
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada; David Braley Research Institute, Hamilton, Ontario, Canada; Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Marc G Jeschke
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada; David Braley Research Institute, Hamilton, Ontario, Canada; Hamilton Health Sciences, Hamilton, Ontario, Canada; Department of Surgery, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
31
|
Fu Y, Zhang J, Qin R, Ren Y, Zhou T, Han B, Liu B. Activating autophagy to eliminate toxic protein aggregates with small molecules in neurodegenerative diseases. Pharmacol Rev 2025; 77:100053. [PMID: 40187044 DOI: 10.1016/j.pharmr.2025.100053] [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: 12/31/2023] [Accepted: 12/05/2024] [Indexed: 04/07/2025] Open
Abstract
Neurodegenerative diseases (NDs), such as Alzheimer disease, Parkinson disease, Huntington disease, amyotrophic lateral sclerosis, and frontotemporal dementia, are well known to pose formidable challenges for their treatment due to their intricate pathogenesis and substantial variability among patients, including differences in environmental exposures and genetic predispositions. One of the defining characteristics of NDs is widely reported to be the buildup of misfolded proteins. For example, Alzheimer disease is marked by amyloid beta and hyperphosphorylated Tau aggregates, whereas Parkinson disease exhibits α-synuclein aggregates. Amyotrophic lateral sclerosis and frontotemporal dementia exhibit TAR DNA-binding protein 43, superoxide dismutase 1, and fused-in sarcoma protein aggregates, and Huntington disease involves mutant huntingtin and polyglutamine aggregates. These misfolded proteins are the key biomarkers of NDs and also serve as potential therapeutic targets, as they can be addressed through autophagy, a process that removes excess cellular inclusions to maintain homeostasis. Various forms of autophagy, including macroautophagy, chaperone-mediated autophagy, and microautophagy, hold a promise in eliminating toxic proteins implicated in NDs. In this review, we focus on elucidating the regulatory connections between autophagy and toxic proteins in NDs, summarizing the cause of the aggregates, exploring their impact on autophagy mechanisms, and discussing how autophagy can regulate toxic protein aggregation. Moreover, we underscore the activation of autophagy as a potential therapeutic strategy across different NDs and small molecules capable of activating autophagy pathways, such as rapamycin targeting the mTOR pathway to clear α-synuclein and Sertraline targeting the AMPK/mTOR/RPS6KB1 pathway to clear Tau, to further illustrate their potential in NDs' therapeutic intervention. Together, these findings would provide new insights into current research trends and propose small-molecule drugs targeting autophagy as promising potential strategies for the future ND therapies. SIGNIFICANCE STATEMENT: This review provides an in-depth overview of the potential of activating autophagy to eliminate toxic protein aggregates in the treatment of neurodegenerative diseases. It also elucidates the fascinating interrelationships between toxic proteins and the process of autophagy of "chasing and escaping" phenomenon. Moreover, the review further discusses the progress utilizing small molecules to activate autophagy to improve the efficacy of therapies for neurodegenerative diseases by removing toxic protein aggregates.
Collapse
Affiliation(s)
- Yuqi Fu
- Institute of Precision Drug Innovation and Cancer Center, the Second Hospital of Dalian Medical University, Dalian, China; Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jin Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China; School of Pharmaceutical Sciences of Medical School, Shenzhen University, Shenzhen, China
| | - Rui Qin
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yueting Ren
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China; Department of Brain Science, Faculty of Medicine, Imperial College, London, UK
| | - Tingting Zhou
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai, China; Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Second Military Medical University, Shanghai, China.
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Bo Liu
- Institute of Precision Drug Innovation and Cancer Center, the Second Hospital of Dalian Medical University, Dalian, China; Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
32
|
Cisa-Wieczorek S, Hernández-Alvarez MI, Parreño M, Muñoz JP, Bussaglia E, Carricondo M, Ubeda J, Dubreuil P, Zorzano A, Brenet F, Nomdedeu JF. D816V KIT mutation induces mitochondrial morphologic and functional changes through BNIP3 downregulation in human myeloid cell lines ROSA and TF-1. Exp Hematol 2025; 145:104748. [PMID: 39986568 DOI: 10.1016/j.exphem.2025.104748] [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: 07/09/2024] [Revised: 02/05/2025] [Accepted: 02/12/2025] [Indexed: 02/24/2025]
Abstract
The KIT receptor is a transmembrane protein found on the surface of many different cell types. Mutant forms of KIT are drivers of myeloid neoplasms, including systemic mastocytosis. The KIT D816V mutation is the most common, leading to constitutive activation of the receptor and its downstream targets, and it is highly resistant to c-KIT inhibitors. Metabolic rewiring is a common trait in cancer. We analyzed the metabolic profile induced by the KIT D816 mutation, measuring mitochondrial parameters in two myeloid cell lines. We found that the KIT D816V mutation causes a significant increase in mitochondrial abundance and activity associated with superoxide production, which could promote DNA instability. Functional and morphologic changes in mitochondria were associated with reduced levels of BNIP3 protein expression. We also detected low BNIP3 levels in clinical acute myeloid leukemia samples harboring D816V mutations. In addition, we have found constitutive mTOR activation in mutated cells, a pathway that has been shown to regulate autophagy. Our data suggest that KIT D816V increases mitochondrial activity through downregulation of BNIP3 expression, which increases mitochondrial number through the autophagy pathway. Alterations in the cellular metabolism induced by the KIT D816V mutation could be therapeutically exploited.
Collapse
Affiliation(s)
- Sabina Cisa-Wieczorek
- Laboratory of Hematology, Department of Hematology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona/IIB Sant Pau, Spain
| | - Maria Isabel Hernández-Alvarez
- Departament de Bioquímica i Biomedicina Molecular, Universitat de Barcelona, Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Matilde Parreño
- Translational Molecular Oncology, IIB Sant Pau, Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain
| | - Juan P Muñoz
- Departament de Bioquímica i Biomedicina Molecular, Universitat de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain; Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Elena Bussaglia
- Laboratory of Hematology, Department of Hematology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona/IIB Sant Pau, Spain
| | - Maite Carricondo
- Laboratory of Hematology, Department of Hematology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona/IIB Sant Pau, Spain
| | - Jose Ubeda
- Laboratory of Hematology, Department of Hematology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona/IIB Sant Pau, Spain
| | - Patrice Dubreuil
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, UMR7258 CNRS, Aix-Marseille Université, Institut Paoli-Calmettes, Marseille, France
| | - Antonio Zorzano
- Departament de Bioquímica i Biomedicina Molecular, Universitat de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain; Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Fabienne Brenet
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, UMR7258 CNRS, Aix-Marseille Université, Institut Paoli-Calmettes, Marseille, France
| | - Josep F Nomdedeu
- Laboratory of Hematology, Department of Hematology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona/IIB Sant Pau, Spain.
| |
Collapse
|
33
|
Wu Y, Yin M, Xia W, Dou B, Liu X, Sun R. Enhancing NK Cell Antitumor Activity With Natural Compounds: Research Advances and Molecular Mechanisms. Phytother Res 2025; 39:1905-1929. [PMID: 39931789 DOI: 10.1002/ptr.8456] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 01/14/2025] [Accepted: 01/25/2025] [Indexed: 05/21/2025]
Abstract
In recent years, immunotherapy has become a novel antitumor strategy in addition to traditional surgery, radiotherapy, and chemotherapy and has exhibited promising results in clinical applications. Despite significant breakthroughs in immunotherapy, such as immune checkpoint blockade and CAR-T cell therapy, it remains necessary to develop more efficacious, safer, and cheaper immunotherapeutic drugs due to factors including small reaction populations, acquired resistance, adverse side effects, and high costs. Natural killer (NK) cells are preeminent cytotoxic lymphocytes of the innate immune system that act as the first line of defense against tumors and synergistically enhance the adaptive immune response of T lymphocytes. Therefore, boosting the antitumor function of NK cells is an important direction in the development of immunotherapy. For decades, various immunotherapies such as adoptive cell therapy, antibody drugs, cytokines supplement, and chemical immunomodulators have been developing rapidly to improve the function of NK cells. Compared to biological immunotherapy, immunomodulators derived from natural products have outstanding advantages of low immunogenicity, multi-targeting, and cost-effectiveness. Currently, increasing attention is being focused on discovering NK cell-stimulating agents from natural products, such as polysaccharides, alkaloids, terpenoids, saponins, phenolics, and quinones. This review aims to categorize and summarize the comprehensive research progress on these natural products, discuss their potential molecular mechanisms in regulating NK cells, and explore their clinical applications as standalone treatments or in combination with conventional chemotherapy regimens.
Collapse
Affiliation(s)
- Yu Wu
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, P. R. China
| | - Mingxiao Yin
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, P. R. China
| | - Wenjiao Xia
- Department of Urology, Center for Oncology Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, P. R. China
| | - Baokai Dou
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, P. R. China
| | - Xiaoyu Liu
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ocean University of China, Qingdao, P. R. China
| | - Ru Sun
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, P. R. China
| |
Collapse
|
34
|
Wang D, Mei Z, Zhao T, Tian H, Peng Z, Kang X, Zhang Y, Qi X. The roles of plant-derived nanovesicles in malignant tumours: A bibliometric analysis. Int J Biol Macromol 2025; 305:141112. [PMID: 39971079 DOI: 10.1016/j.ijbiomac.2025.141112] [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: 12/25/2023] [Revised: 01/09/2025] [Accepted: 02/13/2025] [Indexed: 02/21/2025]
Abstract
Malignant tumours remain one of the most intractable health problems worldwide. Recently, plant-derived nanovesicles (PDNVs) have emerged as a promising tool in the treatment of malignant tumours, leveraging their high biosafety and potential mechanisms such as cancer-selective apoptosis induction and cell cycle arrest. This paper presents a systematic review of the research progress of nanovesicles in malignant tumours, with a focus on plant-derived vesicles (PDVs) and their potential applications in cancer treatment, based on bibliometric analysis. In this review, the research on PDNVs in malignant tumours was identified and analysed through various countries/institutions, authors, references and research hotspots. Furthermore, we summarized the diverse biological functions and applications of PDNVs sourced from various origins in malignant tumours, both when acting independently and as carriers. Lastly, we provide an outlook on the potential applications of PDNVs in malignant tumours. The purpose of this paper is to summarize the research progress of the role of PDNVs in malignant tumours, and to provide new ideas and clues for overcoming the difficulties of tumour treatment.
Collapse
Affiliation(s)
- Dandan Wang
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, China
| | - Zifan Mei
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, China
| | - Tingting Zhao
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, China
| | - Hao Tian
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, China
| | - Zaihui Peng
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, China
| | - Xia Kang
- Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command, Chengdu, Sichuan, China.
| | - Yi Zhang
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, China.
| | - Xiaowei Qi
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
35
|
Xiao J, Kang X, Li N, Hu J, Wang Y, Si J, Pan Y, Zhang S. The role of the poly(A) binding protein-binding protein MoPbp1 as a regulator of the TOR signaling pathway in growth, autophagy, and pathogenicity of the rice blast fungus. Int J Biol Macromol 2025; 306:141730. [PMID: 40043978 DOI: 10.1016/j.ijbiomac.2025.141730] [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: 11/28/2024] [Revised: 03/01/2025] [Accepted: 03/02/2025] [Indexed: 05/11/2025]
Abstract
The target of the rapamycin (TOR) signaling pathway is crucial for biological function in plant pathogenic fungi, yet its regulatory mechanisms remain limited. In this study, the biological functions of MoPbp1 were identified and characterized, and the findings indicate that MoPbp1 contributes to hyphal growth, conidiation, appressoria formation, metabolism of glycogen and lipid droplets, responses to stress, and pathogenicity in Magnaporthe oryzae. Further investigation revealed that MoPBP1 acts as a negative regulator of TOR activity and influences autophagy. In addition, transcriptome data revealed that MoPBP1 mainly regulates amino acid metabolism pathways, components of membrane, and oxidation-reduction process. Our results suggest that MoPbp1 is required for autophagy and pathogenicity in M. oryzae. Overall, we first revealed the relationship between Pbp1 and TOR activity in plant pathogenic fungi.
Collapse
Affiliation(s)
- Junlian Xiao
- Department of Plant Pathology, College of Plant Protection, Anhui Agricultural University, Hefei 230036, China; Anhui Province Key Laboratory of Crop Integrated Pest Management, Anhui Agricultural University, Hefei 230036, China
| | - Xiaoru Kang
- Department of Plant Pathology, College of Plant Protection, Anhui Agricultural University, Hefei 230036, China; Anhui Province Key Laboratory of Crop Integrated Pest Management, Anhui Agricultural University, Hefei 230036, China
| | - Na Li
- Department of Plant Pathology, College of Plant Protection, Anhui Agricultural University, Hefei 230036, China; Anhui Province Key Laboratory of Crop Integrated Pest Management, Anhui Agricultural University, Hefei 230036, China
| | - Jinmei Hu
- Department of Plant Pathology, College of Plant Protection, Anhui Agricultural University, Hefei 230036, China; Anhui Province Key Laboratory of Crop Integrated Pest Management, Anhui Agricultural University, Hefei 230036, China
| | - Yu Wang
- Department of Plant Pathology, College of Plant Protection, Anhui Agricultural University, Hefei 230036, China; Anhui Province Key Laboratory of Crop Integrated Pest Management, Anhui Agricultural University, Hefei 230036, China
| | - Jianyu Si
- Department of Plant Pathology, College of Plant Protection, Anhui Agricultural University, Hefei 230036, China; Anhui Province Key Laboratory of Crop Integrated Pest Management, Anhui Agricultural University, Hefei 230036, China
| | - Yuemin Pan
- Department of Plant Pathology, College of Plant Protection, Anhui Agricultural University, Hefei 230036, China; Anhui Province Key Laboratory of Crop Integrated Pest Management, Anhui Agricultural University, Hefei 230036, China.
| | - Shulin Zhang
- Department of Plant Pathology, College of Plant Protection, Anhui Agricultural University, Hefei 230036, China; Anhui Province Key Laboratory of Crop Integrated Pest Management, Anhui Agricultural University, Hefei 230036, China.
| |
Collapse
|
36
|
Zaater MA, El Kerdawy AM, Mahmoud WR, Abou-Seri SM. Going beyond ATP binding site as a novel inhibitor design strategy for tau protein kinases in the treatment of Alzheimer's disease: A review. Int J Biol Macromol 2025; 307:142141. [PMID: 40090653 DOI: 10.1016/j.ijbiomac.2025.142141] [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: 12/16/2024] [Revised: 03/01/2025] [Accepted: 03/13/2025] [Indexed: 03/18/2025]
Abstract
Alzheimer's disease (AD) is among the top mortality causing diseases worldwide. The presence of extracellular β-amyloidosis, as well as intraneuronal neurofibrillary aggregates of the abnormally hyperphosphorylated tau protein are two major characteristics of AD. Targeting protein kinases that are involved in the disease pathways has been a common approach in the fight against AD. Unfortunately, most kinase inhibitors currently available target the adenosine triphosphate (ATP)- binding site, which has proven unsuccessful due to issues with selectivity and resistance. As a result, a pressing need to find other alternative sites beyond the ATP- binding site has profoundly evolved. In this review, we will showcase some case studies of inhibitors of tau protein kinases acting beyond ATP binding site which have shown promising results in alleviating AD.
Collapse
Affiliation(s)
- Marwa A Zaater
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Kasr El Aini Street, Cairo 11562, Egypt
| | - Ahmed M El Kerdawy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Kasr El Aini Street, Cairo 11562, Egypt; School of Health and Care Sciences, College of Health and Science, University of Lincoln, Joseph Banks Laboratories, Green Lane, Lincoln, United Kingdom.
| | - Walaa R Mahmoud
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Kasr El Aini Street, Cairo 11562, Egypt
| | - Sahar M Abou-Seri
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Kasr El Aini Street, Cairo 11562, Egypt
| |
Collapse
|
37
|
Zhang W, Jia X, Lou D, Wu Q, Yan Y, Mao F. Comparison of mTOR inhibitors combined with endocrine therapy versus that alone in breast cancer: a meta-analysis. Future Oncol 2025; 21:1417-1427. [PMID: 40152674 PMCID: PMC12051556 DOI: 10.1080/14796694.2025.2485022] [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: 01/10/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND This meta-analysis aims to evaluate the efficacy and safety of rapamycin (mTOR) inhibitors with endocrine therapy versus endocrine therapy alone in treating advanced or metastatic estrogen receptor/progesterone receptor (ER/PR) + breast cancer. METHODS We conducted a comprehensive search in PubMed, Web of Science, Embase, and the Cochrane Library for randomized controlled trials (RCTs) comparing mTOR inhibitors plus endocrine therapy with endocrine therapy alone up to September 2024. RESULTS This analysis included 10 RCTs comprising 3,337 patients. Relative to endocrine therapy alone, the combination of mTOR inhibitors and endocrine therapy significantly improved the clinical benefit rate (RR = 1.41, p < 0.001), overall response rate (RR = 1.40, p = 0.006), progression-free survival (PFS; HR = 0.67, p < 0.001), and overall survival (OS; HR = 0.86, p = 0.056), although the improvement in OS was not statistically significant. Subgroup analyses indicated a more pronounced PFS advantage in patients under 65 years of age (HR = 0.55, p = 0.013) and those who had previously received chemotherapy (HR = 0.51, p = 0.001). However, the incidence of adverse events was higher in the combination therapy group, notably stomatitis (p < 0.001), elevated aspartate aminotransferase/alanine aminotransferase (p = 0.04), and diarrhea (p = 0.01). CONCLUSIONS The combination of mTOR inhibitors with endocrine therapy offers superior efficacy with manageable toxicities in patients with advanced or metastatic ER/PR+ breast cancer.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Breast Surgery, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
| | - Xinru Jia
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Dandi Lou
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Qingping Wu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Yici Yan
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Feiyan Mao
- Department of Breast Surgery, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
| |
Collapse
|
38
|
Hamadmad S, Heisler‐Taylor T, Goswami S, Hawthorn E, Chaurasia S, Martini D, Summitt D, Zatari A, Shalash R, Sohail M, Urbanski EG, Bernstein K, Racine J, Satoskar A, El‐Hodiri HM, Fischer AJ, Cebulla CM. Ibudilast Protects Retinal Bipolar Cells From Excitotoxic Retinal Damage and Activates the mTOR Pathway. Glia 2025; 73:905-927. [PMID: 39916387 PMCID: PMC11920683 DOI: 10.1002/glia.24657] [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: 03/04/2024] [Revised: 10/09/2024] [Accepted: 11/24/2024] [Indexed: 02/12/2025]
Abstract
Ibudilast, an inhibitor of macrophage migration inhibitory factor (MIF) and phosphodiesterase (PDE), has been recently shown to have neuroprotective effects in a variety of neurologic diseases. We utilize a chick excitotoxic retinal damage model to investigate ibudilast's potential to protect retinal neurons. Using single cell RNA-sequencing (scRNA-seq), we find that MIF, putative MIF receptors CD74 and CD44, and several PDEs are upregulated in different retinal cells during damage. Intravitreal ibudilast is well tolerated in the eye and causes no evidence of toxicity. Ibudilast effectively protects neurons in the inner nuclear layer from NMDA-induced cell death, restores retinal layer thickness on spectral domain optical coherence tomography (SD-OCT), and preserves retinal neuron function, particularly for the ON bipolar cells, as assessed by electroretinography. PDE inhibition seems essential for ibudilast's neuroprotection, as AV1013, the analogue that lacks PDE inhibitor activity, is ineffective. scRNA-seq analysis reveals upregulation of multiple signaling pathways, including mTOR, in damaged Müller glia (MG) with ibudilast treatment compared to AV1013. Components of mTORC1 and mTORC2 are upregulated in both bipolar cells and MG with ibudilast. The mTOR inhibitor rapamycin blocked accumulation of pS6 but did not reduce TUNEL positive dying cells. Additionally, through ligand-receptor interaction analysis, crosstalk between bipolar cells and MG may be important for neuroprotection. We have identified several paracrine signaling pathways that are known to contribute to cell survival and neuroprotection and might play essential roles in ibudilast function. These findings highlight ibudilast's potential to protect inner retinal neurons during damage and show promise for future clinical translation.
Collapse
Affiliation(s)
- Sumaya Hamadmad
- Department of Ophthalmology and Visual Sciences, Havener Eye InstituteThe Ohio State University, Wexner Medical CenterColumbusOhioUSA
| | - Tyler Heisler‐Taylor
- Department of Ophthalmology and Visual Sciences, Havener Eye InstituteThe Ohio State University, Wexner Medical CenterColumbusOhioUSA
| | - Sandeep Goswami
- Department of Ophthalmology and Visual Sciences, Havener Eye InstituteThe Ohio State University, Wexner Medical CenterColumbusOhioUSA
| | - Evan Hawthorn
- Department of Ophthalmology and Visual Sciences, Havener Eye InstituteThe Ohio State University, Wexner Medical CenterColumbusOhioUSA
| | - Sameer Chaurasia
- Department of Ophthalmology and Visual Sciences, Havener Eye InstituteThe Ohio State University, Wexner Medical CenterColumbusOhioUSA
| | - Dena Martini
- Department of Ophthalmology and Visual Sciences, Havener Eye InstituteThe Ohio State University, Wexner Medical CenterColumbusOhioUSA
| | - Diana Summitt
- Department of Ophthalmology and Visual Sciences, Havener Eye InstituteThe Ohio State University, Wexner Medical CenterColumbusOhioUSA
| | - Ali Zatari
- Department of Ophthalmology and Visual Sciences, Havener Eye InstituteThe Ohio State University, Wexner Medical CenterColumbusOhioUSA
| | - Rahaf Shalash
- Department of Ophthalmology and Visual Sciences, Havener Eye InstituteThe Ohio State University, Wexner Medical CenterColumbusOhioUSA
| | - Misha Sohail
- Department of Ophthalmology and Visual Sciences, Havener Eye InstituteThe Ohio State University, Wexner Medical CenterColumbusOhioUSA
| | - Elizabeth G. Urbanski
- Department of Ophthalmology and Visual Sciences, Havener Eye InstituteThe Ohio State University, Wexner Medical CenterColumbusOhioUSA
| | - Kayla Bernstein
- Department of Ophthalmology and Visual Sciences, Havener Eye InstituteThe Ohio State University, Wexner Medical CenterColumbusOhioUSA
| | | | - Abhay Satoskar
- Department of PathologyThe Ohio State University, Wexner Medical CenterColumbusOhioUSA
| | - Heithem M. El‐Hodiri
- Department of Neuroscience, College of MedicineThe Ohio State UniversityColumbusOhioUSA
| | - Andy J. Fischer
- Department of Neuroscience, College of MedicineThe Ohio State UniversityColumbusOhioUSA
| | - Colleen M. Cebulla
- Department of Ophthalmology and Visual Sciences, Havener Eye InstituteThe Ohio State University, Wexner Medical CenterColumbusOhioUSA
| |
Collapse
|
39
|
Li S, Li Z, Wang J, Han X, Zhang L. Cardamom synergizes with cisplatin against human osteosarcoma cells by mTOR-mediated autophagy. Cancer Gene Ther 2025; 32:538-549. [PMID: 40140723 PMCID: PMC12086087 DOI: 10.1038/s41417-025-00894-9] [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: 10/25/2024] [Revised: 02/21/2025] [Accepted: 03/14/2025] [Indexed: 03/28/2025]
Abstract
Cisplatin (DDP), a frontline chemotherapeutic agent in osteosarcoma (OS) treatment, is frequently paired with other compounds to enhance its therapeutic potency. Cardamom (CAR), a natural flavonoid, exhibits significant inhibitory effects on human OS cells while minimizing toxic side effects. In this study, we combined CAR and DDP to treat OS, revealing that the DDP/CAR combination synergistically inhibits the growth of human OS cells in vitro and in vivo. Network pharmacological analysis indicated that mammalian target of rapamycin (mTOR) may be an important cross-target for DDP/CAR combination. Notably, this combined treatment significantly reduced mTOR phosphorylation and elevated autophagy levels within OS cells. At the mechanistic level, the DDP/CAR regimen enhanced apoptosis and compromised the viability of OS cells by triggering autophagy. This impact was attenuated by the use of the mTOR activator MHY and the autophagy inhibitor hydroxychloroquine (HCQ). Furthermore, in DDP-resistant cell lines, CAR was able to mitigate DDP resistance by bolstering autophagy levels. In general, our results suggest that CAR bolstering autophagy levels DDP against OS cells through the induction of mTOR-mediated autophagy.
Collapse
Affiliation(s)
- Sheng Li
- Department of Respiratory Medicine, Shapingba Hospital affiliated to Chongqing University, Chongqing, China
| | - Ziyun Li
- Key Laboratory of Clinical Laboratory Diagnostics Designated By Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Jiayu Wang
- Key Laboratory of Clinical Laboratory Diagnostics Designated By Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Xueqian Han
- Key Laboratory of Clinical Laboratory Diagnostics Designated By Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Lulu Zhang
- Department of Clinical Laboratory, Medical Sciences Research Center, University-Town Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
40
|
Wan X, Wang D. Curcumin: Epigenetic Modulation and Tumor Immunity in Antitumor Therapy. PLANTA MEDICA 2025; 91:320-337. [PMID: 39689889 DOI: 10.1055/a-2499-1140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
Curcumin (turmeric) is the main ingredient of the Chinese herbal turmeric rhizome, used to treat tumors, diabetes, inflammation, neurodegenerative diseases, cardiovascular diseases, metabolic syndrome, and liver diseases. The antitumor effects of curcumin have received even more attention. One of the main mechanisms of the antitumor effects includes inhibition of tumor invasion and migration, induction of tumor cell apoptosis, and inhibition of various cell signaling pathways. It has been found that the antitumor biological activity of curcumin in the body is associated with epigenetic mechanisms. That also implies that curcumin may act as a potential epigenetic modulator to influence the development of tumor diseases. The immune system plays an essential role in the development of tumorigenesis. Tumor immunotherapy is currently one of the most promising research directions in the field of tumor therapy. Curcumin has been found to have significant regulatory effects on tumor immunity and is expected to be a novel adjuvant for tumor immunity. This paper summarizes the antitumor effects of curcumin from four aspects: molecular and epigenetic mechanisms of curcumin against a tumor, mechanisms of curcumin modulation of tumor immunotherapy, reversal of chemotherapy resistance, and a novel drug delivery system of curcumin, which provide new directions for the development of new antitumor drugs.
Collapse
Affiliation(s)
- Xin Wan
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Dong Wang
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
41
|
Kalenta H, Kilroe SP, Romsdahl TB, Marchant ED, Maroto R, Linares JJ, Russell WK, Rasmussen BB. Constitutively active mTORC1 signaling modifies the skeletal muscle metabolome and lipidome response to exercise. J Appl Physiol (1985) 2025; 138:1173-1186. [PMID: 40215109 DOI: 10.1152/japplphysiol.00987.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/21/2025] [Accepted: 04/04/2025] [Indexed: 05/01/2025] Open
Abstract
A chronic increase in the Mammalian Target of Rapamycin Complex 1 (mTORC1) signaling is implicated in reduced longevity, altered metabolism, and mitochondrial dysfunction. Abnormal mTORC1 signaling may also be involved in the etiology of sarcopenia. To better understand the role of mTORC1 signaling in the regulation of muscle metabolism, we developed an inducible muscle-specific knockout model of DEP domain-containing 5 protein (DEPDC5 mKO), which results in constitutively active mTORC1 signaling. We hypothesized that constitutively active mTORC1 signaling in skeletal muscle would alter the metabolomic and lipidomic response to an acute bout of exercise. Wild-type (WT) and DEPDC5 muscle-specific knockout (KO) mice were studied at rest and following a 1 h bout of treadmill exercise. Acute exercise induced an increased reliance on glycolytic and pentose phosphate pathway (PPP) metabolites in the muscle of mice with hyperactive mTORC1. Lipidomic analysis showed an increase in triglycerides (TGs) in KO mice. Although exercise had a pronounced effect on muscle metabolism, the genotype effect was larger, indicating that constitutively active mTORC1 signaling exerts a dominant influence on metabolic and lipidomic regulation. We conclude that increased mTORC1 signaling shifts muscle metabolism toward greater reliance on nonoxidative energy sources in response to exercise. Understanding the mechanisms responsible for these effects may lead to the development of strategies for restoring proper mTORC1 signaling in conditions such as aging and sarcopenia.NEW & NOTEWORTHY This study demonstrates that hyperactive mTORC1 alters the muscle metabolomic and lipidomic response to exercise, with genotype having a larger effect than exercise. Knockout mice exhibited an increase in reliance on glycolysis and pentose phosphate pathway and an increase in triglyceride turnover. Wild-type mice primarily showed an increase in utilization of TCA cycle and lipid metabolism intermediates.
Collapse
Affiliation(s)
- Hanna Kalenta
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States
| | - Sean P Kilroe
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States
| | - Trevor B Romsdahl
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, United States
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States
| | - Erik D Marchant
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States
| | - Rosario Maroto
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, United States
| | - Jennifer J Linares
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, United States
- Mass Spectrometry Facility, University of Texas Medical Branch, Galveston, Texas, United States
| | - William K Russell
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, United States
- Mass Spectrometry Facility, University of Texas Medical Branch, Galveston, Texas, United States
| | - Blake B Rasmussen
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States
| |
Collapse
|
42
|
Bai Y, Xi Y, Gui C, Huang G, Zhou G. Genetic Evidence Supporting a Causal Association Between mTORC1-Dependent Circulating Protein Levels and Diabetic Retinopathy. Transl Vis Sci Technol 2025; 14:4. [PMID: 40314641 PMCID: PMC12054711 DOI: 10.1167/tvst.14.5.4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 04/06/2025] [Indexed: 05/03/2025] Open
Abstract
Purpose The mechanistic target of rapamycin (mTOR) signaling pathway is essential for the onset and progression of diabetic retinopathy (DR). Nevertheless, the impact of mTORC1 downstream proteins in DR remains uncertain. Therefore, we performed a Mendelian randomization (MR) research to assess the causal effect of downstream mTORC1 proteins on DR risk. Methods Summary statistics on mTORC1 downstream proteins and DR were obtained from the INTERVAL and FinnGen studies (14,584 patients and 176,010 controls), respectively. We used various MR techniques, including inverse-variance-weighted, weighted median, and MR-Egger. Possible pleiotropy and heterogeneity were identified through sensitivity analysis. Results Genetically predicted eIF4E was positively correlated to DR risk (odds ratio = 1.057; 95% confidence interval, 1.008-1.109; P = 0.022]. No relationship has been shown for circulating RP-S6K, eIF4G, eIF4A, eIF4E-BP and eIF4B levels with DR formation. There was no heterogeneity or unbalanced level pleiotropy identified. Conclusions Higher levels of serum eIF4E promote the progression of DR, proposing that pharmacological inhibition of eIF4E activity may be a prospective DR therapeutic strategy. Translational Relevance The present study has highlighted the role of eIF4E in the development of DR, establishing the foundation for basic research into DR targets.
Collapse
Affiliation(s)
- Yaqi Bai
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yujia Xi
- The Second Hospital of Shanxi Medical University, Department of Urology, Taiyuan, China
| | - Chenwei Gui
- Department of Ophthalmology, Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Guohai Huang
- Department of Ophthalmology, Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Guohong Zhou
- Department of Ophthalmology, Shanxi Eye Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
43
|
He T, Wang ZY, Xu B, Zhong CJ, Wang LN, Shi HC, Yang ZY, Zhou SQ, Li H, Hu B, Zhu XD, Shen YH, Zhou J, Fan J, Sun HC, Huang C. CXCL6 Reshapes Lipid Metabolism and Induces Neutrophil Extracellular Trap Formation in Cholangiocarcinoma Progression and Immunotherapy Resistance. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2503009. [PMID: 40305734 DOI: 10.1002/advs.202503009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/31/2025] [Indexed: 05/02/2025]
Abstract
The chemokine CXCL6 is identified as a pivotal regulator of biological processes across multiple malignancies. However, its function in cholangiocarcinoma (CCA) is underexplored. Tumor profiling for CXCL6 is performed using a public database. Both in vitro and in vivo experiments are utilized to evaluate the oncogenic effects of CXCL6 on CCA. Additionally, RNA-Seq is employed to detect transcriptomic changes related to CXCL6 expression in CCA cells and neutrophils. Molecular docking, fluorescence colocalization, and Co-IP are used to elucidate a direct interaction between JAKs and CXCR1/2. Additionally, LC-MS lipidomics and explored the impact of CXCL6 on immunotherapy in vivo. CXCL6 is upregulated in CCA tissues and promoted the proliferation and metastasis of CCA. Mechanistically, CXCL6 regulated the CXCR1/2-JAK-STAT/PI3K axis in CCA via autocrine signaling, leading to lipid metabolic reprogramming, and promoted neutrophil extracellular traps (NETs) formation by activating the RAS/MAPK pathway in neutrophils. Eventually, NETs formation induced immunotherapy resistance in CCA by blocking CD8+T cell infiltration. CXCL6 modulates CCA progression through the CXCR1/2-JAK-STAT/PI3K axis and reshaping its lipid metabolism. CXCL6 also mediates immunotherapy resistance through NETs, which may be a potential therapeutic target and biomarker for CCA.
Collapse
Affiliation(s)
- Tian He
- Department of Hepatobiliary Surgery and Liver Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zi-Yi Wang
- Department of Hepatobiliary Surgery and Liver Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Bin Xu
- Department of Hepatobiliary Surgery and Liver Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Cheng-Jie Zhong
- Department of Hepatobiliary Surgery and Liver Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Lu-Na Wang
- Department of Hepatobiliary Surgery and Liver Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Huan-Chen Shi
- Department of Hepatobiliary Surgery and Liver Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zi-Yue Yang
- Department of Hepatobiliary Surgery and Liver Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Shi-Qi Zhou
- Department of Hepatobiliary Surgery and Liver Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Hui Li
- Department of Hepatobiliary Surgery and Liver Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Bo Hu
- Department of Hepatobiliary Surgery and Liver Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xiao-Dong Zhu
- Department of Hepatobiliary Surgery and Liver Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Ying-Hao Shen
- Department of Hepatobiliary Surgery and Liver Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jian Zhou
- Department of Hepatobiliary Surgery and Liver Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jia Fan
- Department of Hepatobiliary Surgery and Liver Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Hui-Chuan Sun
- Department of Hepatobiliary Surgery and Liver Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Cheng Huang
- Department of Hepatobiliary Surgery and Liver Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| |
Collapse
|
44
|
Seo MK, Kim H, Choi AJ, Seog DH, Kho WG, Park SW, Lee JG. Effects of tianeptine on mTORC1-mediated neuronal autophagy in primary rat hippocampal neurons under nutrient deprivation. Sci Rep 2025; 15:14488. [PMID: 40280952 PMCID: PMC12032415 DOI: 10.1038/s41598-025-92988-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 03/04/2025] [Indexed: 04/29/2025] Open
Abstract
The aim of this study was to investigate the effects of the antidepressant tianeptine on the mechanistic target of rapamycin complex 1(mTORC1)-mediated autophagy pathway in primary hippocampal neurons exposed to B27-deprived conditions. When primary hippocampal neurons were treated with tianeptine at doses of 1, 10, 50, and 100 µM for 3 days under B27-deprived conditions, we observed that it activated autophagy and increased the formation of autophagosomes through the upregulation of autophagic proteins, including autophagy-activating kinase 1 (ULK1), Beclin 1, LC3B-II/I, and p62. And at a concentration of 100 µM tianeptine, the decrease in mTORC1 phosphorylation induced by B27 deprivation was significantly reversed. Changes in the expression of autophagic proteins induced by B27 deprivation were reversed by tianeptine treatment in a concentration-dependent manner, and tianeptine significantly reduced the increase in LC3B membrane number induced by B27 deprivation, an effect that was blocked by pretreatment with rapamycin. In conclusion, tianeptine attenuated the activity of mTORC1-mediated autophagy in primary rat hippocampal neurons under B27-deprived conditions. These results may suggest a novel mechanism by which tianeptine may affect autophagy in neurons.
Collapse
Affiliation(s)
- Mi Kyoung Seo
- Paik Institute for Clinical Research, Inje University, Busan, 47392, Republic of Korea
- Department of Convergence Biomedical Science, College of Medicine, Inje University, Busan, 47392, Republic of Korea
| | - Hyewon Kim
- Department of Psychiatry, College of Medicine, Haeundae Paik Hospital, Inje University, Busan, 48108, Republic of Korea
| | - Ah Jeong Choi
- Paik Institute for Clinical Research, Inje University, Busan, 47392, Republic of Korea
| | - Dae-Hyun Seog
- Department of Convergence Biomedical Science, College of Medicine, Inje University, Busan, 47392, Republic of Korea
- Department of Biochemistry, College of Medicine, Inje University, Busan, 47392, Republic of Korea
- Dementia and Neurodegenerative Disease Research Center, College of Medicine, Inje University, Busan, 47392, Republic of Korea
| | - Weon-Gyu Kho
- Paik Institute for Clinical Research, Inje University, Busan, 47392, Republic of Korea
- Department of Parasitology, College of Medicine, Inje University, Busan, 47392, Republic of Korea
| | - Sung Woo Park
- Paik Institute for Clinical Research, Inje University, Busan, 47392, Republic of Korea.
- Department of Convergence Biomedical Science, College of Medicine, Inje University, Busan, 47392, Republic of Korea.
| | - Jung Goo Lee
- Paik Institute for Clinical Research, Inje University, Busan, 47392, Republic of Korea.
- Department of Psychiatry, College of Medicine, Haeundae Paik Hospital, Inje University, Busan, 48108, Republic of Korea.
| |
Collapse
|
45
|
Chai R, Zheng K, Xu T, Wang H, Cheng X, Lu C, Kang Y. SNX10 Is Involved in Ovarian Cancer Cell Metastasis by Repolarizing Tumor-Associated Macrophages Through mTOR1/Lysosomes Pathway. Biomedicines 2025; 13:1021. [PMID: 40426851 DOI: 10.3390/biomedicines13051021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 04/05/2025] [Accepted: 04/15/2025] [Indexed: 05/29/2025] Open
Abstract
Background: Tumor-associated macrophages (TAMs) are prevalent in advanced ovarian cancer tissues and ascites, significantly influencing disease prognosis. However, the mechanisms driving TAM polarization and their tumor-promoting effects remain poorly understood. Methods: The subcellular distribution of SNX10 in ovarian cancer tissues was analyzed using single-cell datasets (GSE147082, GSE58937). The Kaplan-Meier Plotter and GEPIA2 databases were used to evaluate SNX10's prognostic relevance. Lentivirus-mediated SNX10 overexpression in THP-1 cells was employed in tumor cell-macrophage co-culture experiments. Transwell assays and flow cytometry assessed SNX10's effects on ovarian cancer cell metastasis and cisplatin-induced apoptosis. RNA sequencing, Western blotting, lysosomal pH detection, lipid droplet staining, and RT-qPCR were performed to explore SNX10's molecular mechanisms in TAM polarization and immune modulation. Results: SNX10 was specifically expressed in TAMs, promoting their polarization into the M2 phenotype. This enhanced the migration and invasion of ovarian cancer cell lines A2780 and A2780/CP70 while reducing cisplatin-induced apoptosis. SNX10 decreased lipid droplet content, downregulated p-mTOR1, and impaired lysosomal function in TAMs. Additionally, SNX10 differentially modulated PD-L1 mRNA expression in platinum-sensitive and platinum-resistant ovarian cancer cells. Conclusions: SNX10 regulates the mTOR1/lysosome pathway in TAMs, influencing lipid metabolism and indirectly modulating ovarian cancer cell metastasis. It also alters PD-L1 mRNA expression, suggesting a role in shaping the tumor immune microenvironment.
Collapse
Affiliation(s)
- Ranran Chai
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China
| | - Kewei Zheng
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Ting Xu
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China
| | - Hui Wang
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China
| | - Xiaobo Cheng
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China
| | - Chong Lu
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China
| | - Yu Kang
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| |
Collapse
|
46
|
Yu QX, Zhang YL, Li SY, Zhen L, Li DZ. Phenotypic Features of Fourteen Fetal Cases With a PTEN Variant. Prenat Diagn 2025. [PMID: 40261085 DOI: 10.1002/pd.6806] [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: 02/04/2025] [Revised: 04/12/2025] [Accepted: 04/13/2025] [Indexed: 04/24/2025]
Abstract
OBJECTIVE To present the prenatal sonographic features, genomic findings, and pregnancy outcomes of fetuses diagnosed in utero with a PTEN variant. METHOD This retrospective study analyzed 14 cases of PTEN variants identified through prenatal ultrasound and subsequently confirmed by genetic testing. Clinical and laboratory data were collected for these cases, encompassing maternal demographics, prenatal sonographic findings, molecular testing results, and pregnancy outcomes. RESULTS A total of 14 fetuses with (likely) pathogenic PTEN variants were included in the study. Five cases exhibited abnormalities on second-trimester ultrasound scans; specifically, three presented with macrocephaly, one showed mild ventriculomegaly, and another had cardiac defects (coarctation of the aorta and ventricular septal defect). Nine additional cases were identified during the third trimester. Among these nine cases, six displayed macrocephaly with or without polyhydramnios and ventriculomegaly; one case had mild ventriculomegaly; another presented only with mild polyhydramnios; and one was characterized by macrosomia. Eleven cases involved de novo variants, whereas three variants were inherited from parents. CONCLUSIONS Macrocephaly emerged as the most prevalent sign observed in utero that prompted genetic detection of PTEN defects. This study encourages obstetricians to enhance their awareness regarding PTEN-related disorders in fetuses presenting with macrocephaly.
Collapse
Affiliation(s)
- Qu-Xia Yu
- Department of Prenatal Diagnosis, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yong-Ling Zhang
- Department of Prenatal Diagnosis, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Si-Yun Li
- Department of Prenatal Diagnosis, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Li Zhen
- Department of Prenatal Diagnosis, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Dong-Zhi Li
- Department of Prenatal Diagnosis, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
47
|
Mohebinejad M, Kazeminasab F, Ghanbari Rad M, Bagheri R, Razi M, Willoughby D, Dutheil F. The Combined Effect of High-Intensity Interval Training and Time-Restricted Feeding on the AKT-IGF-1-mTOR Signaling Pathway in the Muscle Tissue of Type 2 Diabetic Rats. Nutrients 2025; 17:1404. [PMID: 40362714 PMCID: PMC12073226 DOI: 10.3390/nu17091404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/16/2025] [Accepted: 04/19/2025] [Indexed: 05/15/2025] Open
Abstract
Background/Objectives: High-intensity interval training (HIIT) and time-restricted feeding (TRF) have shown potential in enhancing glucose metabolism, increasing insulin sensitivity, and promoting muscle health. This study investigates the combined effects of HIIT and TRF on the AKT-IGF-1-mTOR signaling pathway in the muscle tissue of type 2 diabetic (T2D) rats. Methods: 42 male Wistar rats (4-5 weeks of age) were included in the study. The animals were randomly divided into two groups: 1. Standard diet (SD) non-diabetic (n = 7) and 2. High-fat diet (HFD n = 35) for 4 weeks. T2D was induced by intraperitoneal injection (IP) of streptozotocin (STZ) at 35 mg/kg. Animals with blood glucose levels ≥ 250 mg/dL were considered diabetic. Diabetic rats were randomly divided into five groups (n = 7): 1. Diabetes-HIIT (D-HIIT), 2. Diabetes-TRF (D-T), 3. Diabetes-combined TRF and HIIT (D-T+HIIT), 4. Diabetes-Untreated Control (D), and 5. Diabetes with metformin (D-MET). The HIIT protocol and TRF regimen were followed for 10 weeks. Muscle tissue was collected for histological analysis, and the expression of proteins related to the AKT-IGF-1-mTOR pathway was measured. Results: Blood glucose levels, insulin resistance (IR), and markers of muscle degradation were significantly improved in the D-T+HIIT and D-MET groups compared to the non-diabetes group. Furthermore, the activation of the AKT and mTOR signaling proteins, as well as increased IGF-1 expression, was significantly elevated in the D-T+HIIT group compared to the diabetic control group and other treatment groups, and approached levels observed in the non-diabetes group. Additionally, muscle fiber size and overall tissue structure were improved in the treatment groups, particularly in the D-T+HIIT group. Conclusions: The combination of HIIT and TRF appears to offer superior benefits in improving muscle protein synthesis, and glucose regulation in T2D rats, as compared to either HIIT or TRF alone. These findings highlight the potential of this combined approach for addressing muscle-related complications in T2D.
Collapse
Affiliation(s)
- Motahareh Mohebinejad
- Department of Physical Education and Sports Science, Faculty of Humanities, University of Kashan, Kashan 87317-53153, Iran;
| | - Fatemeh Kazeminasab
- Department of Physical Education and Sports Science, Faculty of Humanities, University of Kashan, Kashan 87317-53153, Iran;
| | - Mahtab Ghanbari Rad
- Gerash Cellular and Molecular Research Center, Gerash University of Medical Sciences, Gerash 58666-74417, Iran;
| | - Reza Bagheri
- Department of Exercise Physiology, University of Isfahan, Isfahan 81746-73441, Iran;
| | - Mazdak Razi
- Division of Comparative Histology and Embryology, Department of Basic Sciences, Faculty of Veterinary Medicine, Urmia University, Urmia 57561-51818, Iran;
| | - Darryn Willoughby
- Department of Education, Innovation, and Technology, Baylor College of Medicine-School of Medicine, Temple, TX 76513, USA;
| | - Fred Dutheil
- Preventive and Occupational Medicine, Université Clermont Auvergne, CNRS, LaPSCo, Physiological and Psychosocial Stress, CHU Clermont-Ferrand, University Hospital of Clermont-Ferrand, Witty Fit, F-63000 Clermont-Ferrand, France;
| |
Collapse
|
48
|
Unlu Y, Stinson EJ, Krakoff J, Piaggi P. Protein oxidation in non-exercising healthy adults under varying dietary conditions: Physiological determinants, effects on fuel partitioning, and implications for body weight regulation. Metabolism 2025; 169:156270. [PMID: 40268049 DOI: 10.1016/j.metabol.2025.156270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 04/07/2025] [Accepted: 04/16/2025] [Indexed: 04/25/2025]
Abstract
BACKGROUND Protein oxidation (PROTOX) typically accounts for the smallest fraction of daily energy expenditure (24hEE) in humans compared to carbohydrate and lipid oxidation. However, inter-individual differences in PROTOX may explain differences in fuel partitioning and body weight change. We aimed to elucidate the physiological determinants of PROTOX under controlled 24-h dietary conditions, including eucaloric feeding, fasting, and overfeeding diets with variable protein content. METHODS Eighty-six weight-stable healthy volunteers with normal glucose regulation (67 M/19F; age: 37 ± 10 years; BMI: 26.7 ± 4.5 kg/m2, body fat by DXA: 29.0 ± 9.8 %) underwent 24hEE measurements by whole-room calorimetry during energy balance (20 % protein, 50 % carbohydrate), different overfeeding diets (200 % of the daily eucaloric requirement), including three normal-protein (20 %) diets (balanced: 50 % carbohydrate; high-carbohydrate: 75 % carbohydrate; high-fat: 60 % fat), low-protein (3 %) and high-protein (30 %), and 24-h fasting in a randomized crossover design. Urine samples were collected during each 24-h dietary intervention for quantification of PROTOX and catecholamine excretion rates by nitrogen excretion and high-performance liquid chromatography, respectively. RESULTS PROTOX during energy balance (mean ± SD: 372 ± 78 kcal/day) was positively associated with protein intake (r = 0.39, p < 0.001), fat free mass (r = 0.35, p < 0.001), but not with fat mass (p = 0.24). Higher PROTOX was associated with higher 24-h urinary norepinephrine (partial r = 0.27, p = 0.01), but not epinephrine (p = 0.48), excretion rates. During normal-protein diets, higher PROTOX was associated with lower lipid oxidation, but showed no association with carbohydrate oxidation. Inter-individual variability in PROTOX did not predict changes in weight or body composition over two years. CONCLUSION Dietary protein content, lean body mass, and sympathetic nervous system activity are key determinants of PROTOX. Although PROTOX did not predict free-living weight gain, increased PROTOX is associated with decreased lipid oxidation, underscoring its role in fuel partitioning and whole-body energy and substrate balance.
Collapse
Affiliation(s)
- Yigit Unlu
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Department of Health and Human Services, Phoenix, AZ, United States of America
| | - Emma J Stinson
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Department of Health and Human Services, Phoenix, AZ, United States of America
| | - Jonathan Krakoff
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Department of Health and Human Services, Phoenix, AZ, United States of America
| | - Paolo Piaggi
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Department of Health and Human Services, Phoenix, AZ, United States of America; Department of Information Engineering, University of Pisa, Pisa, Italy.
| |
Collapse
|
49
|
Chen M, Jin J, Bi H, Zhang Y, Sun M, Li X, Wang Y. Advances in the study of NMDA receptors in depression pathogenesis and the antidepressant efficacy of their antagonists. Asian J Psychiatr 2025; 108:104502. [PMID: 40300235 DOI: 10.1016/j.ajp.2025.104502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 04/13/2025] [Accepted: 04/14/2025] [Indexed: 05/01/2025]
Abstract
N-methyl-D-aspartate receptors (NMDA receptors) play a crucial role as ionotropic glutamate receptors in regulating neuroplasticity, learning, memory, and a range of psychiatric disorders. Studies indicate that dysfunction of NMDA receptors is a key pathological mechanism in depression, where abnormal activation can result in neuronal excitotoxicity, excessive extracellular calcium ion accumulation, and disrupted neuroplasticity. As a non-competitive NMDA receptor antagonist, ketamine quickly relieves depressive symptoms by decreasing the activity of extracellular NMDA receptors and activating the mTOR signaling pathway. The treatment can improve severe depression and suicide thoughts within hours, but its potential for hallucinations, dissociative symptoms, and dependency restricts its broader application. Esketamine has demonstrated improvements in both side effects and efficacy and has received FDA approval, while other compounds with NMDA receptor modulating functions, such as memantine and rapastinel, are also showing potential in exploration. Future studies should concentrate on the molecular mechanisms of NMDA receptors, aiming to develop safer and more effective medications, and refine treatment strategies to offer personalized choices and longer-lasting efficacy for the treatment of depression.
Collapse
Affiliation(s)
- Mingrui Chen
- Department of Psychiatry, The First Hospital of China Medical University, China
| | - Jingyan Jin
- Department of Psychiatry, The First Hospital of China Medical University, China
| | - Hongsheng Bi
- Department of Psychiatry, The First Hospital of China Medical University, China; The third hospital of Daqing, Psychiatric Ward No. 9, China
| | - Yihan Zhang
- Department of Psychiatry, The First Hospital of China Medical University, China
| | - Mingyuan Sun
- Department of Psychiatry, The First Hospital of China Medical University, China
| | - Xiaobai Li
- Department of Psychiatry, The First Hospital of China Medical University, China.
| | - Yan Wang
- Center for Psychological Development, China Medical University, China.
| |
Collapse
|
50
|
Zhang C, Guo J. Cell cycle disorders in podocytes: an emerging and increasingly recognized phenomenon. Cell Death Discov 2025; 11:182. [PMID: 40246828 PMCID: PMC12006314 DOI: 10.1038/s41420-025-02486-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 04/01/2025] [Accepted: 04/08/2025] [Indexed: 04/19/2025] Open
Abstract
Proteinuria is observed in various kidney diseases and is frequently associated with a compromised glomerular filtration barrier. Podocytes, as a crucial component of this barrier, play an essential role in preserving the kidney's normal filtration function. Podocytes are terminally differentiated cells that typically do not proliferate. However, certain harmful stimuli can trigger podocytes to re-enter the cell cycle. Due to its unique cytoskeletal structure, podocytes are unable to maintain the structure of the foot process and complete cell division at the same time, eventually form binucleated or multinucleated podocytes. Studies have found that podocytes re-entering the cell cycle are more susceptible to injury, and are prone to detachment from the basement membrane or apoptosis, which are accompanied by the widening of foot processes. This eventually leads to podocyte mitotic catastrophe and the development of proteinuria. Podocyte cell cycle disorders have previously been found mainly in focal segmental glomerulosclerosis and IgA nephropathy. In recent years, this phenomenon has been frequently identified in diabetic kidney disease and lupus nephritis. An expanding body of research has begun to investigate the mechanisms underlying podocyte cell cycle disorders, including cell cycle re-entry, cell cycle arrest, and mitotic catastrophe. This review consolidates the existing literature on podocyte cell cycle disorders in renal diseases and summarizes the molecules that trigger podocyte re-entry into the cell cycle, thereby providing new drug targets for mitigating podocyte damage. This is essential for alleviating podocyte injury, reducing proteinuria, and delaying the progression of kidney diseases.
Collapse
Affiliation(s)
- Chaojie Zhang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, Henan, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Jia Guo
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, Henan, China.
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|