1
|
Albassam H, Almutairi O, Alnasser M, Altowairqi F, Almutairi F, Alobid S. Discovery of a selective PI3Kα inhibitor via structure-based virtual screening for targeted colorectal cancer therapy. J Enzyme Inhib Med Chem 2025; 40:2468852. [PMID: 39992303 PMCID: PMC11852364 DOI: 10.1080/14756366.2025.2468852] [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: 10/24/2024] [Revised: 01/31/2025] [Accepted: 02/08/2025] [Indexed: 02/25/2025] Open
Abstract
Colorectal cancer (CRC) remains a leading cause of cancer-related mortality globally, driving an urgent need for effective therapies. A promising avenue of research focuses on the PI3K/AKT/mTOR signalling pathway, which is frequently disrupted by mutations in the PI3Kα subunit. Our cutting-edge study employed a structure-based virtual screening of ∼3000 compounds, leading to the discovery of F0608-0019, a highly potent and selective PI3Kα inhibitor. F0608-0019 demonstrated remarkable efficacy in suppressing HCT116 colorectal cancer cell proliferation, with an IC50 of 12.14 µM, while maintaining high selectivity by minimising activity against other PI3K isoforms. Advanced molecular dynamics simulations highlighted the stability of F0608-0019's binding interactions with key amino acids, such as TRP:780, ILE:932, and VAL:850, which are critical for its targeted action. These exciting findings reveal F0608-0019 as a leading candidate for innovative CRC therapies that selectively target PI3Kα dysregulation, offering promising new possibilities for effective CRC treatment.
Collapse
Affiliation(s)
- Hussam Albassam
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Omar Almutairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Majed Alnasser
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Faisal Altowairqi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Faris Almutairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Saad Alobid
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
2
|
Merz LM, Winter K, Richter S, Kallendrusch S, Horn A, Grunewald S, Klöting N, Krause K, Kiess W, Le Duc D, Garten A. Effects of alpelisib treatment on murine Pten-deficient lipomas. Adipocyte 2025; 14:2468275. [PMID: 39962643 PMCID: PMC11844927 DOI: 10.1080/21623945.2025.2468275] [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: 09/25/2024] [Revised: 01/20/2025] [Accepted: 01/31/2025] [Indexed: 02/23/2025] Open
Abstract
Phosphatase and tensin homolog (PTEN) hamartoma tumour syndrome (PHTS) is a rare disorder caused by germline mutations in the tumour suppressor gene PTEN, a key negative regulator of phosphatidylinositol 3-kinase (PI3K)/AKT signalling. Children with PHTS often develop lipomas, for which only surgical resection is available as treatment. We investigated the effects of the selective PI3K-inhibitor alpelisib on Pten-deficient lipomas. After incubation with alpelisib or the non-selective PI3K inhibitor wortmannin, we analysed histology, gene expression, and Pi3k pathway in lipoma and control epididymal adipose tissue (epiWAT). Alpelisib increased adipocyte area in lipomas compared to epiWAT. Baseline gene expression showed higher levels of markers for proliferation (Pcna), fibrosis (Tgfb1), and adipogenesis (Pparg) in lipomas, while hormone-sensitive lipase expression was lower than in epiWAT. Following alpelisib incubation, target genes of Pi3k signalling and extracellular matrix factors were reduced. We confirmed Pi3k inhibition through detecting decreased Akt levels compared to control treatment. Human lipoma samples treated with alpelisib showed variable lipolysis responses, suggesting variability in therapeutic outcomes. We established an ex vivo model to study alpelisib effects on Pten-deficient lipomas. These results underscore the therapeutic potential of targeted PI3K inhibition in the treatment of PHTS-associated lipomas, particularly in cases that are inoperable.
Collapse
Affiliation(s)
- Lea M. Merz
- Center for Pediatric Research, University Hospital for Children & Adolescents, Leipzig University, Leipzig, Germany
| | - Karsten Winter
- Institute of Anatomy, Leipzig University, Leipzig, Germany
| | - Sandy Richter
- Center for Pediatric Research, University Hospital for Children & Adolescents, Leipzig University, Leipzig, Germany
| | - Sonja Kallendrusch
- Institute of Anatomy, Leipzig University, Leipzig, Germany
- Institute of Clinical Research and Systems Medicine, Health and Medical University Potsdam, Potsdam, Germany
| | - Andreas Horn
- Institute of Anatomy, Leipzig University, Leipzig, Germany
| | - Sonja Grunewald
- Department for Dermatology, Venereology and Allergology, University Hospital Leipzig, Leipzig, Germany
| | - Nora Klöting
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG), Helmholtz Center Munich at the University and University Hospital Leipzig, Leipzig, Germany
| | - Kerstin Krause
- Department of Endocrinology, Nephrology and Rheumatology, University Hospital Leipzig, Leipzig, Germany
| | - Wieland Kiess
- Center for Pediatric Research, University Hospital for Children & Adolescents, Leipzig University, Leipzig, Germany
| | - Diana Le Duc
- Institute of Human Genetics, University Hospital Leipzig, Leipzig, Germany
| | - Antje Garten
- Center for Pediatric Research, University Hospital for Children & Adolescents, Leipzig University, Leipzig, Germany
| |
Collapse
|
3
|
Kumari S, Akhter M, Gupta GD, Sharma K. Progression and expansion of ALK inhibitors against NSCLC: A dual target approach. Eur J Med Chem 2025; 293:117722. [PMID: 40339471 DOI: 10.1016/j.ejmech.2025.117722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/19/2025] [Accepted: 05/02/2025] [Indexed: 05/10/2025]
Abstract
ALK gene is a member of the tyrosine kinase receptor family found on chromosome 2 (2p23) that plays an important role in the progression of the non-small cell lung cancer (NSCLC). Since the ALK inhibitors such as Crizotinib, Ceritinib, Brigatinib, Alectinib and Lorlatinib, was endorsed for the treatment of advanced NSCLC linked to ALK gene rearrangement. But eventually, patients become resistant to the medication, which will result in treatment failure. However, treatment for NSCLC could be greatly advanced by the development of dual inhibitors that target ALK in addition to other oncogenic pathways like ROS1, c-MET, EGFR, etc. These strategies seek to improve therapy efficacy, address resistance mechanisms, and provide treatment alternatives for patients with intricate molecular profiles. The aim of this review is to summarize the introduction to ALK and the synergy between ALK and other anti-tumor targets, recent developments in the synthesis of various dual inhibitors of the ALK. We also thoroughly discussed their design concepts, structure-activity relationships (SARs), preclinical and clinical data as well as in silico studies to provide ideas for further development of novel ALK based dual inhibitors.
Collapse
Affiliation(s)
- Shreya Kumari
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Mymoona Akhter
- Department of Pharmaceutical Chemistry, SPER, Jamia Hamdard, New Delhi, 110062, India
| | - Ghanshyam Das Gupta
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Kalicharan Sharma
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab, 142001, India.
| |
Collapse
|
4
|
Yan Y, Li M, Guo L, Zhang W, Wu R, Guan T, Ling J, Yang Y, Liu M, Gu X, Liu Y. Silk fibroin hydrogel with recombinant silk fibroin/NT3 protein enhances wound healing by promoting type III collagen synthesis and hair follicle regeneration in skin injury. Mater Today Bio 2025; 33:101957. [PMID: 40575657 PMCID: PMC12198047 DOI: 10.1016/j.mtbio.2025.101957] [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: 02/19/2025] [Revised: 05/08/2025] [Accepted: 06/06/2025] [Indexed: 06/29/2025] Open
Abstract
Scar formation on skin wounds remains a considerable challenge in regenerative medicine. Various wound dressings, composed of biomaterials alone or in combination with bioactive factors, have been developed to improve healing outcomes. In this study, we designed a recombinant neurotrophin-3 (NT3) containing a silk fibroin light chain (SFL) and developed a silk fibroin (SF) hydrogel with NT3 activity. The SFL-NT3 protein bound to the heavy-light chains of SF and was efficiently integrated into the SF hydrogel. We evaluated the effect of the recombinant NT3-SFL hydrogel on wound healing in a mouse skin injury model. This hydrogel enhanced wound healing. Remarkably, SFL-NT3 increased the levels of type III collagen (Col3) during the healing process and induced hair follicle formation, which is a characteristic of scar-less healing. Further investigation revealed that SFL-NT3 upregulated Col3 expression in skin fibroblasts expressing the NT3 receptor, TrkC. NT3 activation of TrkC leads to Akt phosphorylation, resulting in elevated Sox2 levels, which in turn enhances Col3 transcription. Notably, TrkC inhibition abrogated the beneficial effects of SF + SFL-NT3 on wound healing, confirming its involvement in this signaling pathway. In conclusion, the SF hydrogel loaded with SFL-NT3 facilitated rapid and reduced scarring during wound healing, providing a promising approach for the clinical treatment of SF-based biomaterials that incorporate bioactive factors.
Collapse
Affiliation(s)
- Yingying Yan
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, and Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, China
| | - Mingxuan Li
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, China
| | - Longyu Guo
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, China
| | - Wenxue Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, China
| | - Ronghua Wu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, China
| | - Tuchen Guan
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, China
| | - Jue Ling
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, China
| | - Yumin Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, China
| | - Mei Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, China
| | - Xiaosong Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, China
| | - Yan Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, China
| |
Collapse
|
5
|
Dou J, Xiao H, Chen Y, Han W, Zhang S, Wu D, Chen S, Ma Y, Cai Z, Luan Q, Cui L. Diesel exhaust promoted diethylnitrosamine-induced hepatocarcinogenesis in mice. JOURNAL OF HAZARDOUS MATERIALS 2025; 492:138219. [PMID: 40220387 DOI: 10.1016/j.jhazmat.2025.138219] [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: 01/06/2025] [Revised: 04/01/2025] [Accepted: 04/07/2025] [Indexed: 04/14/2025]
Abstract
Exposure to diesel exhaust (DE) has been linked to an increased risk of various cancers, including liver cancer. However, the underlying mechanisms driving this association remain insufficiently understood. In this study, we employed a diethylnitrosamine (DEN)-induced mouse liver tumor model and conducted a 19-week combined exposure (750 μg/m3) using a DE exposure system. Our results demonstrated that long-term DE exposure activates cancer-related genes and enhances the formation of DEN-induced liver tumors. Compared to the DEN group, mice in the DEN + diesel exhaust exposure (DEE) group exhibited lower body weight, higher tumor formation rates and more severe DNA damage. The tumor-promoting effect of DE may be associated with the upregulation of SEMA4D and the activation of the PI3K/AKT signaling pathway. Additionally, liver cells in the DEE group exhibited nuclear atypia, a characteristic feature of cancerous transformation. In vitro studies have revealed that exposure to diesel exhaust particles (DEP) promotes the proliferation of HepG2 cells and HUH7 cells by upregulating SEMA4D and activating the PI3K/AKT signaling pathway. This effect was attenuated by inhibiting either SEMA4D or PI3K. This study was the first to identify that DE exposure promotes the development of DEN-induced liver tumors in mice, with the mechanism potentially involving the SEMA4D/PI3K/AKT pathway. These findings provide novel insights into the hepatotoxic effects of DE and highlight the need for further investigation into its carcinogenic potential.
Collapse
Affiliation(s)
- Junjie Dou
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao, China
| | - Hua Xiao
- Department of Occupational disease, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| | - Yixin Chen
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao, China
| | - Wei Han
- Department of General Practice, Qingdao Key Laboratory of Common Diseases, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Shuxin Zhang
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao, China
| | - Dong Wu
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao, China
| | - Sixin Chen
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao, China
| | - Yuanyuan Ma
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao, China
| | - Zhengguo Cai
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao, China
| | - Qi Luan
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao, China
| | - Lianhua Cui
- Department of Occupational and Environmental Health, School of Public Health, Qingdao University, Qingdao, China.
| |
Collapse
|
6
|
Wu W, Wang X, Ma R, Huang S, Li H, Lyu X. Deciphering the roles of neddylation modification in hepatocellular carcinoma: Molecular mechanisms and targeted therapeutics. Genes Dis 2025; 12:101483. [PMID: 40290125 PMCID: PMC12022649 DOI: 10.1016/j.gendis.2024.101483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 08/05/2024] [Accepted: 11/02/2024] [Indexed: 04/30/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is the most prevalent type of malignant liver tumor with high morbidity and mortality and severely threatens human health and life quality. Thus, it is of great significance to investigate the molecular mechanism underlying the pathogenesis of HCC and seek biomarkers for early diagnosis. Neddylation, one of the most conserved post-translational modification types in eukaryotes, plays vital roles in the progression of HCC. During the process of neddylation, NEDD8 is covalently conjugated to its substrate proteins, thereby modulating multiple necessary biological processes. Currently, increasing evidence shows that the aberrant activation of neddylation is positively correlated with the occurrence and development of tumors and the poor clinical prognosis of HCC patients. Based on the current investigations, neddylation modification has been reported to target both the cullins and non-cullin substrates and subsequently affect HCC progression, including the virus infection, malignant transformation, tumor cell proliferation, migration and invasion ability, and tumor microenvironment. Therefore, inhibitors targeting the neddylation cascade have been developed and entered clinical trials, indicating satisfactory anti-HCC treatment effects. This review aims to summarize the latest progress in the molecular mechanism of pathologically aberrant neddylation in HCC, as well as the advances of neddylation-targeted inhibitors as potential drugs for HCC treatment.
Collapse
Affiliation(s)
- Wenxin Wu
- School of Clinical and Basic Medical Sciences, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250117, China
| | - Xuanyi Wang
- School of Clinical and Basic Medical Sciences, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250117, China
| | - Ruijie Ma
- Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Shuhong Huang
- School of Clinical and Basic Medical Sciences, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250117, China
- Science and Technology Innovation Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250117, China
| | - Hongguang Li
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Xinxing Lyu
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong 250117, China
- School of Clinical and Basic Medical Sciences, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250117, China
- Science and Technology Innovation Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250117, China
| |
Collapse
|
7
|
Weissig V, Joshi MD, Migrino RQ. Cytoprotective effects of liposomal ganglioside GM1. J Liposome Res 2025; 35:212-217. [PMID: 39827412 DOI: 10.1080/08982104.2025.2451776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 12/09/2024] [Accepted: 01/06/2025] [Indexed: 01/22/2025]
Abstract
Gangliosides, glycosphingolipids with one or more N-acetyl-neuraminic acid groups, play essential roles in various cellular and biological processes, among them are cell signaling, neuronal development, cell-cell recognition and the modulation of immune response. Based on their multiple biological roles, the pharmacological utilization of gangliosides for the therapy of several clinical conditions is currently widely being explored but hampered by its limited water solubility. To increase the bioavailability of poorly water-soluble therapeutic agents, pharmaceutical nanocarriers such as liposomes have been developed over the last fifty years. Ganglioside GM1 incorporated into liposomes was proposed during the 1980s for rendering them long-circulating following their intravenous administration, but GM1 was soon replaced by polyethylene glycol which gave rise to the concept of Stealth Liposomes. More recently, the ability of exogenous GM1 to ameliorate oxidative stress was revealed, leading us to investigate the cytoprotective effect of liposomal GM1 under a variety of pathological conditions. Here we review all data showing the antioxidant effect of exogeneous GM1 and based on literature findings and our own, we propose a mechanism by which liposomal exogenous GM1 is able to trigger the Nrf2 (Nuclear factor erythroid 2-related factor 2) pathway, which is a critical cellular defense mechanism protecting against oxidative stress and other types of cellular damage.
Collapse
Affiliation(s)
- Volkmar Weissig
- College of Pharmacy Glendale, Department of Pharmaceutical Sciences, Midwestern University, Glendale, Arizona, USA
| | - Medha D Joshi
- College of Pharmacy Glendale, Department of Pharmaceutical Sciences, Midwestern University, Glendale, Arizona, USA
| | - Raymond Q Migrino
- Phoenix Veterans Affairs Healthcare System, Phoenix, Arizona, USA
- University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, USA
| |
Collapse
|
8
|
Gotkine M, Schoenfeld DA, Cohen I, Shefner JM, Lerner Y, Cohen IR, Klein C, Ovadia E, Cudkowicz ME, the Pooled Resource Open‐Access ALS Clinical Trials Consortium. Akt Activation With IPL344 Treatment for Amyotrophic Lateral Sclerosis: First in Human, Open-Label Study. Muscle Nerve 2025; 71:1032-1042. [PMID: 40105198 PMCID: PMC12060625 DOI: 10.1002/mus.28393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/26/2025] [Accepted: 02/28/2025] [Indexed: 03/20/2025]
Abstract
INTRODUCTION/AIMS Akt intracellular signal transduction pathway dysfunction has been reported in people with amyotrophic lateral sclerosis (ALS) providing a novel target for intervention in this devastating progressive disease. This first-in-human study evaluated the safety, tolerability, and preliminary efficacy of the Akt pathway activator, IPL344, in people with ALS. METHODS Nine participants with ALS and a progression rate > 0.55 points/month on the Amyotrophic Lateral Sclerosis Functional Rating Scale (ALSFRS-R) received open-label IPL344 treatment (once-daily) for up to 36 months. Safety was assessed through adverse event (AE) reporting. Plasma neurofilament light chain (NfL) concentrations were measured before and after treatment. Clinical outcomes were compared to historical data. RESULTS The mean ± SD duration of IPL344 follow-up was 14.0 ± 12.5 months. One participant developed drug hypersensitivity, two had central venous catheter-related AEs, and two had serious pneumonia AEs. The unadjusted mean ± SE slope of decline in ALSFRS-R was -0.53 ± 0.15 (48% slower progression vs. historical controls, p = 0.028). Adjustment for disease stage and rate-indicating covariates indicated a 64% slower ALSFRS-R progression (p = 0.034), with increased rather than reduced body weight (p = 0.02). Eight of nine IPL344-treated participants had a significantly improved slope compared to the median slope of a matched control group (p = 0.04). Plasma NfL concentrations were lowered by 27% (n = 6). Unadjusted median survival for participants in the IPL344 group was 43.4 months [95% CI: 20.5, NA] compared with 19.1 months [17.4, 23.0] in the historical control group. DISCUSSION These preliminary data indicate that IPL344 was safe and well-tolerated, and possibly effective. Our findings may merit further investigation in a larger placebo-controlled clinical trial.
Collapse
Affiliation(s)
- Marc Gotkine
- Neuromuscular Unit, Department of NeurologyHadassah Medical Organization and Faculty of Medicine, Hebrew University of JerusalemJerusalemIsrael
| | - David A. Schoenfeld
- Department of BiostatisticsMassachusetts General HospitalBostonMassachusettsUSA
| | | | - Jeremy M. Shefner
- Department of NeurologyBarrow Neurological InstitutePhoenixArizonaUSA
| | - Yossef Lerner
- Neuromuscular Unit, Department of NeurologyHadassah Medical Organization and Faculty of Medicine, Hebrew University of JerusalemJerusalemIsrael
| | - Irun R. Cohen
- Department of Immunology and Regenerative BiologyWeizmann Institute of ScienceRehovotIsrael
| | - Colin Klein
- Department of NeurologyMeir Medical CenterKfar SabaIsrael
| | | | - Merit E. Cudkowicz
- Department of NeurologyHealey & AMG Center for ALS, Massachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | | |
Collapse
|
9
|
Cetiz MV, Ahmed S, Zengin G, Sinan KI, Emre G, Dolina K, Kalyniukova A, Uba AI, Koyuncu I, Yuksekdag O, Li MY. Bioinformatic and experimental approaches to uncover the bio-potential of Mercurialis annua extracts based on chemical constituents. J Mol Liq 2025; 427:127390. [DOI: 10.1016/j.molliq.2025.127390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2025]
|
10
|
He J, Wong LY, Chen S, Zhang SJ, Chen W, Bai JX, Wang L, Wang XQ, Li SMA, Li Q, Fu XQ, Yu ZL. Inhibition of the PI3K/AKT signaling pathway contributes to the anti-renal cell carcinoma effects of deoxyelephantopin. Biomed Pharmacother 2025; 187:118136. [PMID: 40344699 DOI: 10.1016/j.biopha.2025.118136] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/17/2025] [Accepted: 05/05/2025] [Indexed: 05/11/2025] Open
Abstract
Renal cell carcinoma (RCC) is the most common kidney cancer. Despite advances in treatment, current therapeutic strategies are often limited by side effects, drug resistance, and low response rates, necessitating alternatives for RCC treatment. Deoxyelephantopin (DEO), a sesquiterpene lactone from Elephantopi Herba, has demonstrated anticancer properties in multiple cancer models; however, its effects on RCC remain unknown. This study aimed to investigate the anti-RCC effects of DEO and its underlying molecular mechanisms. Human RCC cell lines (786-O, Caki-1, A498) and a murine RCC cell line (RENCA) were used for in vitro assays. Results revealed that DEO dose-dependently inhibited cell viability and colony formation in 786-O, Caki-1, A498, and RENCA cells, while also inducing apoptosis in 786-O and Caki-1 cells. A RENCA allograft mouse model was used for in vivo assays. DEO significantly suppressed tumor growth without causing notable changes in body weight, organ coefficients, or serum biochemical markers (ALT, AST, BUN, Cr). Network pharmacology analysis predicted the PI3K/AKT signaling pathway as a key mediator of DEO's anti-RCC effects. Western blotting showed that DEO downregulated the expression of EGFR, p-EGFR (Tyr1068), PI3K p110α, p-Akt (Ser473), mTOR, p-mTOR (Ser2448), p-p70S6K (Thr389), 4E-BP1, p-4E-BP1 (Thr37/46), HIF-1α, and Bcl-2. Overactivation of AKT attenuated DEO's inhibitory effects on cell viability in 786-O cells. In conclusion, this study is the first to demonstrate that DEO exerts anti-RCC effects in both cellular and animal models, primarily through inhibition of the PI3K/AKT pathway. These findings suggest that DEO holds promise as a lead compound for RCC management.
Collapse
Affiliation(s)
- Jinjin He
- Consun Chinese Medicines Research Centre for Renal Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Lut Yi Wong
- Consun Chinese Medicines Research Centre for Renal Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Si Chen
- Consun Chinese Medicines Research Centre for Renal Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong; Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Shi-Jia Zhang
- Consun Chinese Medicines Research Centre for Renal Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong; Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Wei Chen
- Consun Chinese Medicines Research Centre for Renal Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Jing-Xuan Bai
- Consun Chinese Medicines Research Centre for Renal Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Li Wang
- Consun Chinese Medicines Research Centre for Renal Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Xiao-Qi Wang
- Consun Chinese Medicines Research Centre for Renal Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Sze-Man Amy Li
- Consun Chinese Medicines Research Centre for Renal Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Qinglin Li
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Xiu-Qiong Fu
- Consun Chinese Medicines Research Centre for Renal Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong.
| | - Zhi-Ling Yu
- Consun Chinese Medicines Research Centre for Renal Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong; Research and Development Centre for Natural Health Products, HKBU Institute for Research and Continuing Education, Shenzhen, China.
| |
Collapse
|
11
|
Meng Y, Jia Y, He Z, Li J, Yuan L. Aryl Hydrocarbon Receptor-Dependent miRNA-382-5p Mediates the Classical Pyroptosis Induced by Foodborne Benzo(a)pyrene through Targeting IκB in Liver. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:12978-12992. [PMID: 40368843 DOI: 10.1021/acs.jafc.5c00511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
Benzo(a)pyrene (BaP), a carcinogen prevalent in high-temperature processed foods, activates the aryl hydrocarbon receptor (AhR) and induces liver pyroptotic injury. MicroRNAs regulate mRNA expression and are involved in BaP toxicity. In this study, we investigated the essential role of microRNA in BaP-induced pyroptosis. In vivo, BaP induces liver pyroptotic injury by activating AhR, which may be attributed to AhR's influence on microRNA expression. The miRNA-382-5p/Akt and miRNA-382-5p/IκB pairs were predicted to post-transcriptionally regulate pyroptosis. In vitro, miRNA-382-5p activates the classical NF-κB/NLRP3/Caspase-1 pyroptosis signaling pathway by targeting and inhibiting the IκB gene. Furthermore, AhR activation by BaP could promote the high expression of miRNA-382-5p, thereby upregulating the NF-κB/NLRP3/Caspase-1 pyroptosis signaling pathway. In summary, we established that the AhR-mediated miR-382-5p/NF-κB/NLRP3/Caspase-1 axis is a key driver of BaP-induced pyroptosis in hepatocytes and elucidated the underlying mechanisms. These findings provide a valuable theoretical basis for considering miRNA-382-5p as a potential target for preventing BaP toxicity.
Collapse
Affiliation(s)
- Yao Meng
- Engineering Research Center of High Value Utilization of Western China Fruit Resources, Key Laboratory of Food Processing Byproducts for Advanced Development and High Value Utilization, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi' an 710119, People' s Republic of China
| | - Yingyu Jia
- Engineering Research Center of High Value Utilization of Western China Fruit Resources, Key Laboratory of Food Processing Byproducts for Advanced Development and High Value Utilization, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi' an 710119, People' s Republic of China
| | - Ziyan He
- Engineering Research Center of High Value Utilization of Western China Fruit Resources, Key Laboratory of Food Processing Byproducts for Advanced Development and High Value Utilization, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi' an 710119, People' s Republic of China
| | - Jianke Li
- Engineering Research Center of High Value Utilization of Western China Fruit Resources, Key Laboratory of Food Processing Byproducts for Advanced Development and High Value Utilization, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi' an 710119, People' s Republic of China
| | - Li Yuan
- Engineering Research Center of High Value Utilization of Western China Fruit Resources, Key Laboratory of Food Processing Byproducts for Advanced Development and High Value Utilization, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi' an 710119, People' s Republic of China
| |
Collapse
|
12
|
Li Y, Man W, Li X, Wu X, Cui Y, Chen S, Li X, Lin Y, Jiang L, Wang Y. Plasmolipin deficiency is essential for HUVECs survival under hypoxic conditions. Cell Death Discov 2025; 11:239. [PMID: 40379643 DOI: 10.1038/s41420-025-02526-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 04/29/2025] [Accepted: 05/07/2025] [Indexed: 05/19/2025] Open
Abstract
This study aims to explore the molecules that affect the survival of Human Umbilical Vein Endothelial Cells (HUVECs) under hypoxia and their mechanisms of action. In hypoxia, plasmolipin (PLLP) was identified through the screening of CRISPR/Cas9 and small guide RNA (sgRNA) library. Functionally, PLLP knockout led to increase cell proliferation, cellular metabolism, tight junction formation, angiogenesis ability, migration and invasion in hypoxic HUVECs. Furthermore, PLLP knockout countered the inhibitory effects of bevacizumab on HUVECs angiogenesis and cell survival in hypoxic conditions. PLLP knockout was found to modulate the survival of HUVECs in hypoxia by enhancing the phosphorylation of AKT and ERK1/2 proteins. In conclusion, inhibiting the expression of PLLP in HUVECs promotes cell survival and maintenance of cellular functions under hypoxic condition. PLLP plays a crucial role in regulating cell survival in hypoxia through the activation of AKT and ERK1/2 pathways. This study identifies novel molecules that affect HUVECs survival under hypoxic conditions and provides a new possibility for future studies on cell survival under hypoxic conditions.
Collapse
Affiliation(s)
- Yanghua Li
- Medical College, Guangxi University, Nanning, China
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China
| | - Weiling Man
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China
| | - Xiang Li
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China
| | - Xiaojie Wu
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China
| | - Yumeng Cui
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China
| | - Shiyun Chen
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China
| | - Xianhong Li
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China
| | - Yanli Lin
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China.
| | - Lihe Jiang
- Medical College, Guangxi University, Nanning, China.
- School of Basic Medical Sciences, Youjiang Medical University for Nationalities, Baise, China.
- Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China.
| | - Youliang Wang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China.
| |
Collapse
|
13
|
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
|
14
|
Fujii W, Yamazaki O, Hirohama D, Kaseda K, Kuribayashi-Okuma E, Tsuji M, Hosoyamada M, Kochi Y, Shibata S. Gene-environment interaction modifies the association between hyperinsulinemia and serum urate levels through SLC22A12. J Clin Invest 2025; 135:e186633. [PMID: 40100301 PMCID: PMC12077893 DOI: 10.1172/jci186633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 03/12/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUNDHyperinsulinemia and insulin resistance often accompany elevated serum urate levels (hyperuricemia), a highly heritable condition that triggers gout; however, the underlying mechanisms are unclear.METHODSWe evaluated the association between the index of hyperinsulinemia and the fractional excretion of urate (FEUA) in 162 outpatients. The underlying mechanisms were investigated through single-cell data analysis and kinase screening combined with cell culture experiments. In 377,358 participants of the UK Biobank (UKBB), we analyzed serum urate, hyperinsulinemia, and salt intake. We also examined gene-environment interactions using single nucleotide variants in SLC22A12, which encodes urate transporter 1 (URAT1).RESULTSThe index of hyperinsulinemia was inversely associated with FEUA independently of other covariates. Mechanistically, URAT1 cell-surface abundance and urate transport activity were regulated by URAT1-Thr408 phosphorylation, which was stimulated by hyperinsulinemia via AKT. Kinase screening and single-cell data analysis revealed that serum and glucocorticoid-regulated kinase 1 (SGK1), induced by high salt, activated the same pathway, increasing URAT1. Arg405 was essential for these kinases to phosphorylate URAT1-Thr408. In UKBB participants, hyperinsulinemia and high salt intake were independently associated with increased serum urate levels. We found that SLC22A12 expression quantitative trait locus (eQTL) rs475688 synergistically enhanced the positive association between serum urate and hyperinsulinemia.CONCLUSIONURAT1 mediates the association between hyperinsulinemia and hyperuricemia. Our data provide evidence for the role of gene-environment interactions in determining serum urate levels, paving the way for personalized management of hyperuricemia.FUNDINGACRO Research Grants of Teikyo University; Japan Society for the Promotion of Science; the Japanese Society of Gout and Uric & Nucleic Acids; Fuji Yakuhin; Nanken-Kyoten; Medical Research Center Initiative for High Depth Omics.
Collapse
Affiliation(s)
- Wataru Fujii
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
- Department of Genomic Function and Diversity, Medical Research Laboratory, Institute for Integrated Research, Institute of Science Tokyo, Tokyo, Japan
| | - Osamu Yamazaki
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Daigoro Hirohama
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Ken Kaseda
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Emiko Kuribayashi-Okuma
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | | | - Makoto Hosoyamada
- Laboratory of Human Physiology and Pathology, Faculty of Pharma-Science, Teikyo University, Tokyo, Japan
| | - Yuta Kochi
- Department of Genomic Function and Diversity, Medical Research Laboratory, Institute for Integrated Research, Institute of Science Tokyo, Tokyo, Japan
| | - Shigeru Shibata
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
- Advanced Comprehensive Research Organization, Teikyo University, Tokyo, Japan
| |
Collapse
|
15
|
Zhang Q, Yang L, Li C, Zhang Y, Li R, Jia F, Wang L, Ma X, Yao K, Tian H, Zhuo C. Exploring the potential antidepressant mechanisms of ibuprofen and celecoxib based on network pharmacology and molecular docking. J Affect Disord 2025; 377:136-147. [PMID: 39986574 DOI: 10.1016/j.jad.2025.02.053] [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: 08/18/2024] [Revised: 02/07/2025] [Accepted: 02/17/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND Evidence has shown that ibuprofen and celecoxib are effective in improving depressive symptoms, but their mechanisms of action are unclear. In this study, we aimed to determine the relationship between these two drugs and depressive disorder (DD) and elucidate potential mechanisms of action. METHODS Relevant targets for ibuprofen, celecoxib, and DD were obtained and screened from multiple online drug and disease public databases. A protein-protein interaction network was obtained. The Centiscape and CytoHubba plug-ins were applied to screen for core targets. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses were performed. Molecular docking was performed to predict the binding of ibuprofen and celecoxib to core targets. Examined the differences in core target protein expression between DD patients (DDs, n = 18) and healthy controls (HCs, n = 16) as a further experimental validation of the network pharmacology results. RESULTS In total, 220 potential targets for ibuprofen and 316 potential targets for celecoxib were identified and associated with DD. The antidepressant effects of both drugs involve many key targets in pathways such as "pathways in cancer" and "neuroactive ligand-receptor interaction," including ALB, BCL2, MAPK3, SRC, STAT3, EGFR, and PPARG. The binding affinity of ALB with ibuprofen is the strongest, and it is connected only by hydrophobic interactions. Celecoxib exhibits higher affinity at multiple targets such as SRC, EGFR, and PPARG, with stronger and more specific intermolecular interactions, including salt bridges and halogen bonds. Clinical trials have found that serum ALB expression in DDs is significantly lower than that in HCs (t = 6.653, p < 0.001), further confirming the potential role of ibuprofen in DD. CONCLUSIONS Ibuprofen and celecoxib primarily exert their antidepressant effects through targets and pathways related to inflammation, neural signaling, and cancer, with celecoxib showing a stronger potential antidepressant effect. The expression difference of the core target ALB between depression and healthy individuals further supports the potential effect of the drug on DD. Our findings propose new treatment strategies, support the link between inflammation and depression, and encourage reassessing existing medications for depression.
Collapse
Affiliation(s)
- Qiuyu Zhang
- Computational Biology and Animal Imaging Center (CBAC), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Mental Health Center, Tianjin 300222, China; Laboratory of Psychiatric-Neuroimaging-Genetic and Co-morbidity (PNGC_Lab), Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Lei Yang
- Computational Biology and Animal Imaging Center (CBAC), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Mental Health Center, Tianjin 300222, China; Laboratory of Psychiatric-Neuroimaging-Genetic and Co-morbidity (PNGC_Lab), Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Chao Li
- Computational Biology and Animal Imaging Center (CBAC), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Mental Health Center, Tianjin 300222, China; Laboratory of Psychiatric-Neuroimaging-Genetic and Co-morbidity (PNGC_Lab), Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Ying Zhang
- Computational Biology and Animal Imaging Center (CBAC), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Mental Health Center, Tianjin 300222, China; Laboratory of Psychiatric-Neuroimaging-Genetic and Co-morbidity (PNGC_Lab), Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Ranli Li
- Computational Biology and Animal Imaging Center (CBAC), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Mental Health Center, Tianjin 300222, China; Laboratory of Psychiatric-Neuroimaging-Genetic and Co-morbidity (PNGC_Lab), Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Feng Jia
- Computational Biology and Animal Imaging Center (CBAC), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Mental Health Center, Tianjin 300222, China; Laboratory of Psychiatric-Neuroimaging-Genetic and Co-morbidity (PNGC_Lab), Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Lina Wang
- Computational Biology and Animal Imaging Center (CBAC), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Mental Health Center, Tianjin 300222, China; Laboratory of Psychiatric-Neuroimaging-Genetic and Co-morbidity (PNGC_Lab), Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Xiaoyan Ma
- Computational Biology and Animal Imaging Center (CBAC), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Mental Health Center, Tianjin 300222, China; Laboratory of Psychiatric-Neuroimaging-Genetic and Co-morbidity (PNGC_Lab), Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Kaifang Yao
- Computational Biology and Animal Imaging Center (CBAC), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Mental Health Center, Tianjin 300222, China; Laboratory of Psychiatric-Neuroimaging-Genetic and Co-morbidity (PNGC_Lab), Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Hongjun Tian
- Animal Imaging Center (AIC) of Tianjin Fourth Center Hospital, Tianjin Medical University Affiliated Tianjin Fourth Center Hospital, Nankai University Affiliated Tianjin Fourth Center Hospital, Tianjin 300140, China
| | - Chuanjun Zhuo
- Computational Biology and Animal Imaging Center (CBAC), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Mental Health Center, Tianjin 300222, China; Laboratory of Psychiatric-Neuroimaging-Genetic and Co-morbidity (PNGC_Lab), Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin 300222, China.
| |
Collapse
|
16
|
Yang X, Zhu J, Tao X, Gao F, Cai Y, Lv Y, Xie S, Xie K, Lan T, Han J, Wu H. Challenges and opportunities for the diverse substrates of SPOP E3 ubiquitin ligase in cancer. Theranostics 2025; 15:6111-6145. [PMID: 40521202 PMCID: PMC12159753 DOI: 10.7150/thno.113356] [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: 03/06/2025] [Accepted: 04/26/2025] [Indexed: 06/18/2025] Open
Abstract
The Speckle-type POZ protein (SPOP), a substrate adaptor of the cullin-RING E3 ligase complex, mediates both the degradation and non-degradative ubiquitination of substrates, which are crucial for regulating various biological functions and cellular processes. Dysregulation of SPOP-mediated ubiquitination has been implicated in several cancers. Emerging evidence suggests that SPOP functions as a double-edged sword: acting as a tumor suppressor in prostate cancer (PCa), hepatocellular carcinoma (HCC), and colorectal cancer (CRC), while potentially serving as an oncoprotein in kidney cancer (KC). Therefore, SPOP's role in tumorigenesis appears to be tissue- or context-dependent. Numerous downstream substrates of SPOP have been identified across various cancers, where they regulate carcinogenesis, metabolic reprogramming, cell death, immune evasion, therapy resistance, and tumor microenvironment (TME) remodeling. However, the definitive role of SPOP in these cancers requires further investigation. A comprehensive understanding of the molecular mechanisms of SPOP in different cancer types will provide new insights into its function in oncogenesis, potentially advancing anti-cancer drug development. Here, we summarize the latest findings on SPOP's functions and structural features, its regulatory mechanisms, the roles of its substrates in various cancers, and SPOP-targeting strategies.
Collapse
Affiliation(s)
- Xiaojuan Yang
- Liver Digital Transformation Research Laboratory, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, P.R. China
- Department of Biotherapy, Cancer Center and State Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiang Zhu
- Liver Digital Transformation Research Laboratory, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, P.R. China
- Breast Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xue Tao
- Liver Digital Transformation Research Laboratory, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, P.R. China
- Liver Transplantation Center, Liver Digital Transformation Research Laboratory, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, P.R. China
| | - Fengwei Gao
- Liver Digital Transformation Research Laboratory, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, P.R. China
- Liver Transplantation Center, Liver Digital Transformation Research Laboratory, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, P.R. China
| | - Yunshi Cai
- Liver Digital Transformation Research Laboratory, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, P.R. China
- Liver Transplantation Center, Liver Digital Transformation Research Laboratory, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, P.R. China
| | - Yinghao Lv
- Liver Digital Transformation Research Laboratory, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, P.R. China
- Liver Transplantation Center, Liver Digital Transformation Research Laboratory, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, P.R. China
| | - Sinan Xie
- Liver Digital Transformation Research Laboratory, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, P.R. China
- Liver Transplantation Center, Liver Digital Transformation Research Laboratory, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, P.R. China
| | - Kunlin Xie
- Liver Digital Transformation Research Laboratory, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, P.R. China
- Liver Transplantation Center, Liver Digital Transformation Research Laboratory, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, P.R. China
| | - Tian Lan
- Liver Digital Transformation Research Laboratory, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, P.R. China
- Liver Transplantation Center, Liver Digital Transformation Research Laboratory, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, P.R. China
| | - Junhong Han
- Department of Biotherapy, Cancer Center and State Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hong Wu
- Liver Digital Transformation Research Laboratory, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, P.R. China
- Liver Transplantation Center, Liver Digital Transformation Research Laboratory, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
17
|
Shi H, Shi J, Wang Z, Zuo H, Guo T, Zheng H, Xiao R, Zhang X, Yang S, Li J. GAS reduced inflammatory responses in activated microglia by regulating the Ccr2/Akt/Gsk-3β pathway. Mol Brain 2025; 18:40. [PMID: 40329396 PMCID: PMC12057146 DOI: 10.1186/s13041-025-01206-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 04/02/2025] [Indexed: 05/08/2025] Open
Abstract
Hypoxic-ischemic brain damage (HIBD) is a significant cause of neonatal death and neurological dysfunction. Following this injury, activated microglia can lead to a series of inflammatory responses. Gastrodin (GAS), a polyphenol extracted from the Chinese herbal medicine Gastrodia elata Blume, has demonstrated antioxidant and anti-inflammatory effects. This study investigated the neuroprotective impact of GAS in HIBD mice model and in BV2 cells subjected to oxygen-glucose deprivation (OGD) treatment. Expression of various members of the Ccr2/Akt/Gsk-3β, including Ccl2, Ccr2, Akt, p-Akt, Gsk-3β, p-Gsk-3β and inflammatory factors TNF-α and IL-1β in activated microglia was assessed by Western blotting, immunofluorescence, and qRT-PCR in HIBD in postnatal mice, and in OGD-induced BV2 microglia in vitro with or without GAS treatment. The present results showed that GAS effectively reduces the expression of Ccl2 and Ccr2, increases the phosphorylation levels of Akt and Gsk-3β, and decreases the expression of the TNF-α and IL-1β. Additionally, we have shown that inhibition of Ccr2 by RS102895 increased the expression of p-Akt and p-Gsk-3β, and attenuate production of proinflammatory mediators in activated microglia. Of note, the expression of p-Akt, p-Gsk-3β, TNF-α and IL-1β remained unchanged after the combination of gastrodin and RS102895. Taken together, we conclude that GAS can play a protective role in reducing the neuroinflammatory response after HIBD. It is suggested that this is mainly through up-regulating the Akt/Gsk-3β signaling pathway via the Ccr2 receptor in the present experimental paradigm.
Collapse
Affiliation(s)
- Haolong Shi
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, 1168 West Chunrong Road, Kunming, Yunnan, 650500, PR China
| | - Jinsha Shi
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, 1168 West Chunrong Road, Kunming, Yunnan, 650500, PR China
| | - Zhao Wang
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, 1168 West Chunrong Road, Kunming, Yunnan, 650500, PR China
| | - Hanjun Zuo
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, 1168 West Chunrong Road, Kunming, Yunnan, 650500, PR China
| | - Tao Guo
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, 1168 West Chunrong Road, Kunming, Yunnan, 650500, PR China
| | - Huixin Zheng
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, 1168 West Chunrong Road, Kunming, Yunnan, 650500, PR China
| | - Rong Xiao
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, 1168 West Chunrong Road, Kunming, Yunnan, 650500, PR China
| | - Xinglin Zhang
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, 1168 West Chunrong Road, Kunming, Yunnan, 650500, PR China
| | - Shuhan Yang
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, 1168 West Chunrong Road, Kunming, Yunnan, 650500, PR China
| | - Juanjuan Li
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, 1168 West Chunrong Road, Kunming, Yunnan, 650500, PR China.
| |
Collapse
|
18
|
Hedayati N, Safari MH, Milasi YE, Kahkesh S, Farahani N, Khoshnazar SM, Dorostgou Z, Alaei E, Alimohammadi M, Rahimzadeh P, Taheriazam A, Hashemi M. Modulation of the PI3K/Akt signaling pathway by resveratrol in cancer: molecular mechanisms and therapeutic opportunity. Discov Oncol 2025; 16:669. [PMID: 40323335 PMCID: PMC12052642 DOI: 10.1007/s12672-025-02471-w] [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: 11/22/2024] [Accepted: 04/23/2025] [Indexed: 05/08/2025] Open
Abstract
The phosphatidylinositol 3-kinase (PI3K)/Akt pathway is a critical intracellular signaling pathway that is pivotal in various cellular functions. It is in senescence, survival, and growth under normal physiological and pathological conditions, including neoplasms. Additionally, this pathway has been recognized as essential for the regulation of the cell cycle. Several previous studies have indicated that the PI3K/Akt signaling pathway can be influenced by various natural products, with resveratrol (3,4',5-trihydroxy-trans-stilbene) being a particularly important phytoalexin polyphenol in this context. This review explores the impact of the PI3K/Akt signaling pathway on the initiation and advancement of various cancerous conditions and the potential of resveratrol to target this signaling mechanism. The review begins by summarizing the anti-tumor capabilities of resveratrol and then emphasizes the significant role of the PI3K/Akt signaling pathway in the progression of multiple malignancies. Finally, we discuss the therapeutic effects of resveratrol on human neoplasms, from brain cancers to gastrointestinal malignancies, through regulation of this signaling cascade.
Collapse
Affiliation(s)
- Neda Hedayati
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohamad Hosein Safari
- Department of Internal Medicine, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Yaser Eshaghi Milasi
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Samaneh Kahkesh
- Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Najma Farahani
- Farhikhtegan Medical Convergent Sciences Research Center, TeMs. C., Islamic Azad University, Tehran, Iran
| | - Seyedeh Mahdieh Khoshnazar
- Gastroenterology and Hepatology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Zahra Dorostgou
- Department of Biochemistry, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
| | - Elmira Alaei
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Payman Rahimzadeh
- Surgical Research Society (SRS), Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergent Sciences Research Center, TeMs. C., Islamic Azad University, Tehran, Iran.
- Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergent Sciences Research Center, TeMs. C., Islamic Azad University, Tehran, Iran.
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
19
|
Aguayo F, Tapia JC, Calaf GM, Muñoz JP, Osorio JC, Guzmán-Venegas M, Moreno-León C, Levican J, Andrade-Madrigal C. The Role of Xenobiotics and Anelloviruses in Colorectal Cancer: Mechanisms and Perspectives. Int J Mol Sci 2025; 26:4354. [PMID: 40362591 PMCID: PMC12072659 DOI: 10.3390/ijms26094354] [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: 02/24/2025] [Revised: 04/29/2025] [Accepted: 05/01/2025] [Indexed: 05/15/2025] Open
Abstract
Xenobiotics are non-natural chemical compounds to which the human population is exposed. Chronic exposure to certain xenobiotics is associated with various diseases, including cancer development. Anelloviruses (AVs), including Torque Teno Virus (TTV), Torque Teno Mini Virus (TTMV), and Torque Teno Midi Virus (TTMDV), are ubiquitous viruses found in the general population. As no disease has been definitively associated with AVs, they are sometimes referred to as "viruses awaiting a disease". This review explores the potential roles of xenobiotics and AVs in colorectal cancer (CRC) development and suggests a potential interplay between them. Evidence suggests an association between certain xenobiotics (like pesticides, cigarette smoke components, and dietary factors) and CRC, while such an association is less clear for AVs. The high prevalence of AVs suggests these infections alone may be insufficient to disrupt homeostasis; thus, additional factors might be required to promote disease, potentially including cancer.
Collapse
Affiliation(s)
- Francisco Aguayo
- Laboratorio de Oncovirología, Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad de Tarapacá, Arica 1000000, Chile; (J.C.O.); (M.G.-V.); (C.M.-L.); (C.A.-M.)
| | - Julio C. Tapia
- Laboratorio de Transformación Celular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Av. Independencia 1027, Santiago 8380453, Chile
| | - Gloria M. Calaf
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica 1000000, Chile;
| | - Juan P. Muñoz
- Laboratorio de Bioquímica, Departamento de Química, Facultad de Ciencias, Universidad de Tarapacá, Arica 1000007, Chile;
| | - Julio C. Osorio
- Laboratorio de Oncovirología, Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad de Tarapacá, Arica 1000000, Chile; (J.C.O.); (M.G.-V.); (C.M.-L.); (C.A.-M.)
| | - Matías Guzmán-Venegas
- Laboratorio de Oncovirología, Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad de Tarapacá, Arica 1000000, Chile; (J.C.O.); (M.G.-V.); (C.M.-L.); (C.A.-M.)
| | - Carolina Moreno-León
- Laboratorio de Oncovirología, Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad de Tarapacá, Arica 1000000, Chile; (J.C.O.); (M.G.-V.); (C.M.-L.); (C.A.-M.)
| | - Jorge Levican
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Cristian Andrade-Madrigal
- Laboratorio de Oncovirología, Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad de Tarapacá, Arica 1000000, Chile; (J.C.O.); (M.G.-V.); (C.M.-L.); (C.A.-M.)
| |
Collapse
|
20
|
Xu Y, Yang Y, Yu X, Han L, Liu S, Zhang N, Yang M. Integrated functional genomics-identified LYRM4 promotes fumarate accumulation and hepatocellular carcinoma progression. Arch Biochem Biophys 2025; 770:110448. [PMID: 40320061 DOI: 10.1016/j.abb.2025.110448] [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: 04/16/2025] [Accepted: 05/01/2025] [Indexed: 05/11/2025]
Abstract
Multiple risk factors for HCC have been identified, however, not all individuals exposed to these factors will develop HCC, suggesting that genetic predisposition also contributes to hepatocarcinogenesis. Despite the identification of numerous single-nucleotide polymorphisms (SNPs) in HCC risk loci and several protein-coding susceptibility genes through genome-wide association studies (GWAS), the potential mechanisms are still not fully understood. In this study, we used The Updated Integrative Functional Genomics Approach (TUIFGA) method to investigate functional causal genetic variants in HCC. We identified one SNP rs399283 associated with HCC risk, which locates in CREB1 binding motif of the LYRM4 intronic enhancer. The rs399283 genetic variation may affect the binding of CREB1 to the enhancer of the oncogene LYRM4 in HCC, leading to allele-specific gene expression changes. Mechanistically, elevated levels of LYRM4 enhance the enzymatic activities of succinate dehydrogenase (SDH), thereby promoting fumarate accumulation in cells and playing a key role in HCC tumorigenesis. Our results offer valuable insights into the genetic complexity of HCC and emphasize the significant potential of fumarate regulation as a novel approach for cancer therapy.
Collapse
Affiliation(s)
- Yuan Xu
- Affiliated Hospital of Shandong Second Medical University, School of Clinical Medicine, Weifang, 261053, Shandong Province, PR China; Shandong Provincial Key Laboratory of Precision Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong Province, PR China
| | - Yanting Yang
- Shandong Provincial Key Laboratory of Precision Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong Province, PR China
| | - Xinyuan Yu
- Shandong Provincial Key Laboratory of Precision Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong Province, PR China; School of Life Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, 271021, Shandong Province, PR China
| | - Linyu Han
- Shandong Provincial Key Laboratory of Precision Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong Province, PR China
| | - Shuqing Liu
- Shandong University Cancer Center, Jinan, 250117, Shandong Province, PR China
| | - Nasha Zhang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, 250117, Shandong Province, PR China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu Province, PR China.
| | - Ming Yang
- Shandong Provincial Key Laboratory of Precision Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong Province, PR China; School of Life Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, 271021, Shandong Province, PR China; Shandong University Cancer Center, Jinan, 250117, Shandong Province, PR China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu Province, PR China.
| |
Collapse
|
21
|
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
|
22
|
Chu X, Sun J, Liang J, Liu W, Xing Z, Li Q, Li Q. Mechanisms of muscle repair after peripheral nerve injury by electrical stimulation combined with blood flow restriction training. SPORTS MEDICINE AND HEALTH SCIENCE 2025; 7:173-184. [PMID: 39991124 PMCID: PMC11846447 DOI: 10.1016/j.smhs.2024.10.002] [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: 06/07/2024] [Revised: 09/20/2024] [Accepted: 10/09/2024] [Indexed: 02/25/2025] Open
Abstract
This review elucidates the impact of electrical stimulation (ES) and blood flow restriction (BFR) training on muscle function. ES induces a transformation in muscle fibers type by rearranging myosin heavy chain isoform patterns. Additionally, it influences muscle protein synthesis and degradation through specific signaling pathways such as protein kinase B/mechanistic target of rapamycin (Akt/mTOR), as well as via autophagy and the ubiquitin-proteasome system, thereby effectively maintaining muscle mass. BFR, on the other hand, restricts muscle blood flow, leading to metabolic products accumulation and localized hypoxia, which not only promotes the recruitment of fast-twitch fibers but also activates the mTOR signaling pathway, enhancing muscle protein synthesis. The combination of ES and BFR synergistically facilitates muscle protein synthesis through the mTOR pathway, thereby accelerating the recovery of muscle function following peripheral nerve injury.
Collapse
Affiliation(s)
- Xiaolei Chu
- Department of Rehabilitation, Tianjin University Tianjin Hospital, Tianjin, China
| | - Jiaojiao Sun
- Tianjin Key, Laboratory of Exercise Physiology and Sports Medicine, Institute of Sport, Exercise and Health, Tianjin University of Sport, Tianjin, China
| | - Jiajia Liang
- Tianjin Key, Laboratory of Exercise Physiology and Sports Medicine, Institute of Sport, Exercise and Health, Tianjin University of Sport, Tianjin, China
| | - Wenjie Liu
- Tianjin Key, Laboratory of Exercise Physiology and Sports Medicine, Institute of Sport, Exercise and Health, Tianjin University of Sport, Tianjin, China
| | - Zheng Xing
- Department of Rehabilitation, Tianjin University Tianjin Hospital, Tianjin, China
| | - Qi Li
- Department of Rehabilitation, Tianjin University Tianjin Hospital, Tianjin, China
| | - Qingwen Li
- Tianjin Key, Laboratory of Exercise Physiology and Sports Medicine, Institute of Sport, Exercise and Health, Tianjin University of Sport, Tianjin, China
| |
Collapse
|
23
|
Xie S, Yang H. SP1 activates AKT3 to facilitate the development of diabetic nephropathy. J Endocrinol Invest 2025; 48:1269-1281. [PMID: 39786707 DOI: 10.1007/s40618-025-02530-7] [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: 09/18/2024] [Accepted: 01/01/2025] [Indexed: 01/12/2025]
Abstract
BACKGROUND Diabetic nephropathy (DN) is a severe complication of diabetes mellitus and has the complex pathogenesis. The previous study reported that protein kinase Bγ (AKT3) was involved in DN progression. Our aim was to explore the detailed mechanisms of AKT3 in DN development. METHODS RT-qPCR was performed to measure the levels of specificity protein 1 (SP1) and AKT3. Mesangial cells were treated with high glucose (30 mM) to form DN cell model in vitro. Western blot was conducted to detect the protein expression of AKT3, SP1, fibrosis-related proteins, and AKT/mTOR pathway-related proteins. Cell proliferation and inflammation were evaluated via MTT, EdU staining, and ELISA assays, respectively. Oxidative stress was determined via measuring ROS and MDA levels. ChIP and dual-luciferase reporter assays were carried out to verify the relationship between SP1 and AKT3. C57BL/6 mice-treated with streptozotocin for 5 days were used to establish DN mouse model in vivo, and HE and Masson staining were conducted to evaluate pathological changes of mouse kidney tissues. RESULTS AKT3 and SP1 were highly expressed in DN kidney tissues and HG-induced mesangial cells. AKT3 depletion could relieve HG treatment-caused cell damage of mesangial cells through repressing cell proliferation, fibrosis, inflammation and oxidative stress. SP1 can bind to the promoter of AKT3 and serve as a translation regulation factor of AKT3. SP1 overexpression worsened HG treatment-caused cell damage of mesangial cells. Moreover, AKT3 upregulation could block the suppressive effects of SP1 depletion on cell proliferation, fibrosis, inflammation and oxidative stress in HG-induced mesangial cells. SP1 depletion reduced AKT3 expression to inactivate the AKT/mTOR pathway in HG-induced mesangial cells. Besides, AKT3 knockdown inhibited the activation of the AKT/mTOR pathway to hamper the development of DN in mice through alleviating fibrosis and inflammation in vivo. CONCLUSION Our results indicated that SP1 activated AKT3 and AKT/mTOR pathway to promote mesangial cell proliferation, fibrosis, inflammation and oxidative stress, thereby facilitating DN development.
Collapse
Affiliation(s)
- Shanshan Xie
- Department of Endocrinology, Nanshi Hospital of Nanyang, No. 130, West Zhongzhou Road, Nanyang, 473065, China
| | - Han Yang
- Department of Endocrinology, Nanshi Hospital of Nanyang, No. 130, West Zhongzhou Road, Nanyang, 473065, China.
| |
Collapse
|
24
|
Newton F, Halachev M, Nguyen L, McKie L, Mill P, Megaw R. Autophagy disruption and mitochondrial stress precede photoreceptor necroptosis in multiple mouse models of inherited retinal disorders. Nat Commun 2025; 16:4024. [PMID: 40301324 PMCID: PMC12041483 DOI: 10.1038/s41467-025-59165-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 04/14/2025] [Indexed: 05/01/2025] Open
Abstract
Inherited retinal diseases (IRDs) are a leading cause of blindness worldwide. One of the greatest barriers to developing treatments for IRDs is the heterogeneity of these disorders, with causative mutations identified in over 280 genes. It is therefore a priority to find therapies applicable to a broad range of genetic causes. To do so requires a greater understanding of the common or overlapping molecular pathways that lead to photoreceptor death in IRDs and the molecular processes through which they converge. Here, we characterise the contribution of different cell death mechanisms to photoreceptor degeneration and loss throughout disease progression in humanised mouse models of IRDs. Using single-cell transcriptomics, we identify common transcriptional signatures in degenerating photoreceptors. Further, we show that in genetically and functionally distinct IRD models, common early defects in autophagy and mitochondrial damage exist, triggering photoreceptor cell death by necroptosis in later disease stages. These results suggest that, regardless of the underlying genetic cause, these pathways likely contribute to cell death in IRDs. These insights provide potential therapeutic targets for novel, gene-agnostic treatments for IRDs applicable to the majority of patients.
Collapse
Affiliation(s)
- Fay Newton
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road, Edinburgh, EH4 2XU, UK
| | - Mihail Halachev
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road, Edinburgh, EH4 2XU, UK
| | - Linda Nguyen
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road, Edinburgh, EH4 2XU, UK
| | - Lisa McKie
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road, Edinburgh, EH4 2XU, UK
| | - Pleasantine Mill
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road, Edinburgh, EH4 2XU, UK
| | - Roly Megaw
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road, Edinburgh, EH4 2XU, UK.
- Princess Alexandra Eye Pavilion, NHS Lothian, Edinburgh, EH3 9HA, UK.
| |
Collapse
|
25
|
Zhao J, Sun Y, Liswaniso S, Wu H, Sun X, Yan C, Qin N, Xu R. The Effect of FSH-Induced Nuclear Exclusion of FOXO3/4 on Granulosa Cell Proliferation and Apoptosis of Hen Ovarian Follicles. Genes (Basel) 2025; 16:500. [PMID: 40428322 PMCID: PMC12111018 DOI: 10.3390/genes16050500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2025] [Revised: 04/23/2025] [Accepted: 04/24/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Follicle stimulating hormone (FSH) is key regulator for follicular development, differentiation, and maturation, and the effects involve various intra follicular factors, such as members of the forkhead box O (FOXO) subfamily. However, the specific role and mechanism of FOXO3 and FOXO4 in growth and development of hen follicles by affecting granulosa cell (GC) division and FSH response function are still unclear. METHOD This study selected GCs from 6-8 mm chicken follicles, and immunofluorescence and Western blot methods were used to detect FSH-induced FOXO3/4 phosphorylation and nuclear exclusion. Quantitative real-time PCR and flow cytometry were used to investigate the regulatory effects of FSH-induced FOXO3/4 phosphorylation and nuclear exclusion on follicular GC proliferation, differentiation, and apoptosis. RESULTS This study found that the level of p-FOXO3/4 protein significantly increased in cells treated with FSH for 12 h, while the expression level of non-phosphorylated FOXO3/4 significantly decreased. After co-treatment with 10 ng/mL Leptomycin B (LMB), FOXO3/4 phosphorylation was effectively prevented. The immunofluorescence results showed that FOXO3 and FOXO4 were originally distributed in the GC nucleus and cytoplasm, whereas they were almost accumulated in cytoplasm when treated with FSH for 12 h. Conversely, FOXO3/4 nuclear translocation was blocked by LMB. Moreover, RT-qPCR and flow cytometry results showed that FSH treatment significantly increased proliferation and differentiation of cells but significantly reduced GCs apoptosis. However, LMB also eliminated these stimulating or inhibitory effects on cell proliferation. CONCLUSION These findings provide new evidence that FSH-induced FOXO3/4 nuclear exclusion promotes GCs proliferation and reduces GCs apoptosis during hen follicular development.
Collapse
Affiliation(s)
- Jinghua Zhao
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China; (J.Z.); (Y.S.); (S.L.); (H.W.); (X.S.); (C.Y.); (N.Q.)
- School of Life Sciences, Changchun Normal University, Changchun 130032, China
| | - Yuhan Sun
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China; (J.Z.); (Y.S.); (S.L.); (H.W.); (X.S.); (C.Y.); (N.Q.)
| | - Simushi Liswaniso
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China; (J.Z.); (Y.S.); (S.L.); (H.W.); (X.S.); (C.Y.); (N.Q.)
| | - Hengsong Wu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China; (J.Z.); (Y.S.); (S.L.); (H.W.); (X.S.); (C.Y.); (N.Q.)
| | - Xue Sun
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China; (J.Z.); (Y.S.); (S.L.); (H.W.); (X.S.); (C.Y.); (N.Q.)
| | - Chunchi Yan
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China; (J.Z.); (Y.S.); (S.L.); (H.W.); (X.S.); (C.Y.); (N.Q.)
| | - Ning Qin
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China; (J.Z.); (Y.S.); (S.L.); (H.W.); (X.S.); (C.Y.); (N.Q.)
| | - Rifu Xu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China; (J.Z.); (Y.S.); (S.L.); (H.W.); (X.S.); (C.Y.); (N.Q.)
| |
Collapse
|
26
|
Chhaing R, Ma Q, Schuh M, Erkan E. Downregulation of Akt induces proximal tubule epithelial cell apoptosis via Foxo-1-BIM pathway in proteinuric states. RESEARCH SQUARE 2025:rs.3.rs-6234375. [PMID: 40313745 PMCID: PMC12045355 DOI: 10.21203/rs.3.rs-6234375/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Proteinuria is a widely utilized surrogate marker in clinical practice for its predictive and prognostic value. The mechanistic link between proteinuria and progression remains elusive. Proximal tubule epithelial cells(PTEC) retrieve albumin in the glomerular filtrate via receptor mediated endocytosis facilitated by megalin-cubilin complex. We reported that cell-survival protein, Akt phosphorylates cargo binding endocytic adaptor protein to megalin, disabled-2(Dab2). We hypothesize that downregulation of Akt signaling as a result of overwhelmed endocytic machinery in albumin overload is linked to PTEC apoptosis in proteinuric states. We show that cell culture and animal model of albumin overload inhibited phosphorylation of Akt in association with apoptosis in PTEC. Chemical inhibition and overexpression of Akt by constitutively active Akt plasmid exacerbated and alleviated apoptosis respectively in response to albumin overload in PTEC. Mouse with targeted inhibition of Akt1 and Akt2 in PTEC (Akt1/2lox/loxSGLT2cre) displayed perturbed albumin endocytosis at baseline. Albumin overload in Akt1/2 lox/lox SGLT2cre mouse led to dephosphorylation and translocation downstream Akt target, Forkhead box O-1 (Foxo1) to nuclei driving transcriptional activation of proapoptotic BIM followed by translocation of proapoptotic Bax and BIM to mitochondria and cytochrome-c to cytosol. In an effort to investigate the role of Akt in progression, we examined kidney biopsy specimens of patients with focal segmental glomerulosclerosis (FSGS) and minimal change disease. Kidney biopsies of patients with FSGS exhibited decreased pSer473-Akt expression in PTEC early in the course of disease, preceding progression to end stage kidney disease. We conclude that downstream dephosphorylation of Foxo and transcriptional activation of BIM and subsequent mitochondrial injury drives apoptosis following Akt downregulation in PTEC albeit inhibition of albumin endocytosis in proteinuric states.
Collapse
Affiliation(s)
| | - Qing Ma
- Cincinnati Children’s Hospital
| | | | | |
Collapse
|
27
|
Huang L, Nguyen ST, Yang Z, Kirschke CP, Prouteau C, Copin MC, Bonneau D, Blanchet O, Mallebranche C, Pellier I, Coutant R, Miot C, Ziegler A. Reduced AKT activation accompanied with high TP53 expression is implicated in the impaired hematogenesis in Ziegler-Huang syndrome and the Znt7 null mice partially recapitulates the human disease linked to pancytopenia. J Trace Elem Med Biol 2025; 89:127658. [PMID: 40286389 DOI: 10.1016/j.jtemb.2025.127658] [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: 02/27/2025] [Revised: 04/10/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Inherited bone marrow failure (IBMF) is a life-threatening condition. Excessive expression of TP53 induces cell cycle arrest and apoptosis of hematopoietic cells in individuals with IBMF. We recently discovered two pathogenic variants, NM_001144884:c.21dup;p.(Asp8ArgfsTer3) and NM_001144884:c.842 + 15 T > C, in ZNT7 associated with IBMF (Ziegler-Huang Syndrome; BMF8). However, the pathophysiologic mechanism of IBMF caused by ZNT7 mutations remained unknown. METHOD We investigated TP53 expression and the activation of its upstream regulator, AKT, in cell lines from affected individuals. We rescued the wild-type phenotype of AKT activation via transduction of wild-type ZNT7 into patient's fibroblasts. We performed fluorescence microscopy to assess co-expression patterns of ZNT7 with hematopoietic cell markers in different human and mouse bone marrow cell types. Finally, we evaluated the hematological features of Znt7 deficient mice. RESULTS The growth of patient's EBV-transformed B (B-EBV) lymphoblasts was impaired. We observed excessive expression of TP53 in the patient's B-EBV lymphoblasts accompanied by a significant decrease in AKT activation. Importantly, overexpression of wild-type ZNT7 in patient's fibroblasts rescued the activation of the AKT pathway by insulin. Additionally, human ZNT7 was expressed in myeloid and lymphoid lineage cells, whereas mouse ZnT7 was mainly expressed in the nucleated hematopoietic cells in the respective bone marrow. Despite these differences, we observed progressive cytopenia in Znt7KO mice, partially recapitulating BMF8 in humans. CONCLUSION Excessive expression of TP53 and down-regulation of AKT activation induced by ZNT7 deficiency might impair cell survival, which may contribute to the pathophysiology of bone marrow failure in affected individuals with BMF8.
Collapse
Affiliation(s)
- Liping Huang
- USDA/ARS/Western Human Nutrition Research Center, 430 West Health Sciences Drive, Davis, CA 95616, USA; Graduate Group of Nutritional Biology, Department of Nutrition, University of California at Davis, One Shields Ave, Davis, CA 95616, USA; Integrative Genetics and Genomics, University of California at Davis, One Shields Ave, Davis, CA 95616, USA.
| | - Steven T Nguyen
- USDA/ARS/Western Human Nutrition Research Center, 430 West Health Sciences Drive, Davis, CA 95616, USA
| | - Zhongyue Yang
- Graduate Group of Nutritional Biology, Department of Nutrition, University of California at Davis, One Shields Ave, Davis, CA 95616, USA
| | - Catherine P Kirschke
- USDA/ARS/Western Human Nutrition Research Center, 430 West Health Sciences Drive, Davis, CA 95616, USA
| | - Clément Prouteau
- Department of Genetics, University Hospital of Angers, 4 rue Larrey, Cedex 9, Angers 49333, France
| | | | - Dominique Bonneau
- Department of Genetics, University Hospital of Angers, 4 rue Larrey, Cedex 9, Angers 49333, France
| | - Odile Blanchet
- Biobank Resource Center, CHU Angers, University Hospital of Angers, 4 rue Larrey, Cedex 9, BB-0033-00038, 49333, France; University Angers Nantes Université CHU Angers INSERM CNRS CRCI2NA SFR ICAT, Cedex 9, Angers 49333, France
| | - Coralie Mallebranche
- University Angers Nantes Université CHU Angers INSERM CNRS CRCI2NA SFR ICAT, Cedex 9, Angers 49333, France
| | - Isabelle Pellier
- University Angers Nantes Université CHU Angers INSERM CNRS CRCI2NA SFR ICAT, Cedex 9, Angers 49333, France
| | - Régis Coutant
- Department of Pediatric Endocrinology, University Hospital of Angers, Cedex 9, Angers 49333, France; Reference Center for Rare Pituitary Diseases, University of Hospital of Angers, Cedex 9, Angers 49333, France
| | - Charline Miot
- University Angers Nantes Université CHU Angers INSERM CNRS CRCI2NA SFR ICAT, Cedex 9, Angers 49333, France; Immunology and Allergology Laboratory, University Hospital of Angers, 4 rue Larrey, Cedex 9, Angers 49333, France
| | - Alban Ziegler
- Department of Genetics, University Hospital of Angers, 4 rue Larrey, Cedex 9, Angers 49333, France; Deparment of Medical Genetics, University Hospital of Toulouse, Toulouse 31100, France.
| |
Collapse
|
28
|
Liu Z, Shi X, Ke T, Yan Z, Xiong L, Tang F. USP7 promotes endothelial activation to aggravate sepsis-induced acute lung injury through PDK1/AKT/NF-κB signaling pathway. Cell Death Discov 2025; 11:183. [PMID: 40246841 PMCID: PMC12006344 DOI: 10.1038/s41420-025-02481-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 04/02/2025] [Accepted: 04/07/2025] [Indexed: 04/19/2025] Open
Abstract
Disruption of the endothelial cell barrier and the subsequent inflammatory response represent a central pathological feature of acute lung injury (ALI). Ubiquitination plays a pivotal role in regulating protein stability, intracellular transport, and enzyme activity, which is typically reversed by deubiquitinating enzymes. Nevertheless, the function of deubiquitinating enzymes in endothelial biology and in ALI remains largely uninvestigated. The present study demonstrates that the expression of USP7 is increased in instances of endothelial inflammation and ALI. The knockdown or inhibition of USP7 using specific inhibitors was observed to significantly reduce the TNF-α-induced inflammatory response of endothelial cells and their adhesion capacity to monocytes. Conversely, the overexpression of USP7 was observed to promote the inflammatory response and adhesion capacity of endothelial cells. The specific inhibitors of USP7 were found to be effective in mitigating acute lung injury induced by LPS. From a mechanistic perspective, our findings indicate that USP7 binds and deubiquitinates PDK1, thereby stabilizing PDK1 and promoting the activity of the inflammatory pathway in endothelial cells. In conclusion, our findings demonstrate the role of a novel USP7-PDK1 signaling axis in regulating TNF-α-induced vascular endothelial injury and reveal that USP7 is a deubiquitylating enzyme of PDK1. These observations suggest that targeting the USP7-PDK1 axis may offer a promising therapeutic strategy for the treatment of acute lung injury.
Collapse
Affiliation(s)
- Zhiyi Liu
- Department of Anesthesiology and Operative Medicine, Medical Center of Anesthesiology and Pain, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Xiaoyun Shi
- Department of Anesthesiology and Operative Medicine, Medical Center of Anesthesiology and Pain, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Tiantian Ke
- Department of Anesthesiology and Operative Medicine, Medical Center of Anesthesiology and Pain, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Zhisu Yan
- Department of Anesthesiology and Operative Medicine, Medical Center of Anesthesiology and Pain, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Lei Xiong
- Department of Anesthesiology and Operative Medicine, Medical Center of Anesthesiology and Pain, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Fang Tang
- Department of Anesthesiology and Operative Medicine, Medical Center of Anesthesiology and Pain, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
29
|
Gaba S, Sahu M, Chauhan N, Jain U. Transforming growth factor alpha: Key insights into physiological role, cancer therapeutics, and biomarker potential (A review). Int J Biol Macromol 2025; 310:143212. [PMID: 40250676 DOI: 10.1016/j.ijbiomac.2025.143212] [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: 01/29/2025] [Revised: 04/01/2025] [Accepted: 04/14/2025] [Indexed: 04/20/2025]
Abstract
Transforming Growth Factor Alpha (TGF-α) is a critical member of the epidermal growth factor (EGF) family and a key regulator of various physiological processes, including cellular proliferation, survival, differentiation, wound repair, and tissue regeneration. Deficiencies or mutations in TGF-α have been associated with impaired tissue development and organ growth, underscoring its critical role in maintaining normal and healthy physiology. Alterations in its levels are frequently implicated in the neoplastic transformation of cells, contributing to cancer development. Several strategies for targeting TGF-α in cancer therapy have been explored, such as the use of antibodies, recombinant proteins, oligonucleotide-mediated interference in ligand synthesis, ligand sequestration via binding proteins, and modulation of the signal transduction pathway. Furthermore, there is growing interest in the potential of TGF-α as a diagnostic or prognostic biomarker for cancer. This review delves into the role of TGF-α in normal physiology and its involvement in carcinogenesis. It highlights therapies targeting TGF-α and explores future directions in targeting TGF-α/EGFR signaling using advancing approaches, including nanoparticle-based drug delivery systems, CRISPR-Cas genome editing tool, PROTAC, and combination therapies. By bringing attention to this molecule, we aim to explore its untapped potential in cancer treatment and inspire further research into its promising applications across related fields. While recent studies highlight the promise of TGF-α as a clinical biomarker, further research is needed to validate its specificity and integration into personalized medicine. By providing a comprehensive overview of TGF-α in both normal and pathological contexts, this review aims to offer new insights into its translational applications in cancer therapeutics and biomarker discovery.
Collapse
Affiliation(s)
- Smriti Gaba
- School of Health Sciences and Technology, UPES, Dehradun 248007, India
| | - Mridul Sahu
- School of Health Sciences and Technology, UPES, Dehradun 248007, India
| | - Nidhi Chauhan
- School of Health Sciences and Technology, UPES, Dehradun 248007, India
| | - Utkarsh Jain
- School of Health Sciences and Technology, UPES, Dehradun 248007, India.
| |
Collapse
|
30
|
Bolat B, Bayraktaroglu C, Degirmenci Z, Cerah E, Sali M, Kolcu E, Bars DN, Aydin C, Abasova F, Alisoy A, Atali HE, Beker MC, Celik U, Beker M. Unraveling the Role of NeuroD2 in Ischemic Pathophysiology: Insight into Neuroprotection Mechanisms Associated with AKT Survival Kinase. Neuromolecular Med 2025; 27:28. [PMID: 40237843 PMCID: PMC12003519 DOI: 10.1007/s12017-025-08852-2] [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: 01/09/2025] [Accepted: 03/29/2025] [Indexed: 04/18/2025]
Abstract
NeuroD2 (ND2), a neuron-specific transcription factor, is essential in neural differentiation and neuroplasticity, yet its regulation under neuronal injury is barely uncovered. Effective treatment strategies for ischemic conditions require extensive knowledge of the signaling pathways and mechanisms underlying ischemic pathophysiology. This study aims to uncover the neuroprotective role of ND2 in ischemia and its interactions with critical signaling pathways implicated in recovery. An in vitro ischemic stroke model oxygen-glucose deprivation (OGD) method was applied to neuro-2A (N2a) cells with lentiviral ND2 (LvND2) overexpression. DNA fragmentation and cell survival assays indicated ND2's neuroprotective and anti-apoptotic effects under OGD conditions. Proteomic profiling and interaction analyses showed that LvND2 regulated the synthesis of cellular signaling, proliferation and cell adhesion-related proteins, such as MAPK3, Mki67, and NCAM. Additionally, a positive correlation was observed between ND2 expression and phosphorylated AKT levels. To investigate the interaction between ND2 and the PI3K/AKT signaling pathway, the pathway was pharmacologically inhibited with Wortmannin 30 min before OGD induction. After 8 h of OGD followed by 16 h of reperfusion, cell survival, DNA fragmentation, and Western blot analyses were performed. LvND2 administration alone increased cellular survival, whereas its combination with Wortmannin resulted in decreased cell survival. Additionally, LvND2 alone reduced the number of TUNEL-positive cells, while its combination with Wortmannin remains non-significant. These findings suggest that ND2 and AKT function in a coordinated manner within the PI3K/AKT survival pathway. ND2 may modulate AKT activity, highlighting its potential as a therapeutic target for addressing ischemic pathophysiology through molecular therapies.
Collapse
Affiliation(s)
- Busenur Bolat
- Department of Medical Biology, Institute of Health Sciences, University of Health Sciences Türkiye, Istanbul, Türkiye
- Department of Physiology, Institute of Health Sciences, Yeditepe University, Istanbul, Türkiye
| | - Cigdem Bayraktaroglu
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Türkiye
| | - Zehra Degirmenci
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Türkiye
| | - Ecem Cerah
- International School of Medicine, University of Health Sciences Türkiye, Istanbul, Türkiye
| | - Mehmet Sali
- International School of Medicine, University of Health Sciences Türkiye, Istanbul, Türkiye
| | - Edanur Kolcu
- International School of Medicine, University of Health Sciences Türkiye, Istanbul, Türkiye
| | - Dila Nur Bars
- International School of Medicine, University of Health Sciences Türkiye, Istanbul, Türkiye
| | - Cemil Aydin
- International School of Medicine, University of Health Sciences Türkiye, Istanbul, Türkiye
| | - Fatima Abasova
- International School of Medicine, University of Health Sciences Türkiye, Istanbul, Türkiye
| | - Abdulla Alisoy
- International School of Medicine, University of Health Sciences Türkiye, Istanbul, Türkiye
| | - Hasan Ege Atali
- International School of Medicine, University of Health Sciences Türkiye, Istanbul, Türkiye
| | - Mustafa Caglar Beker
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Türkiye
- Department of Physiology, School of Medicine, Istanbul Medeniyet University, Istanbul, Türkiye
| | - Ulkan Celik
- Department of Medical Biology, School of Medicine, University of Health Sciences Türkiye, Istanbul, Türkiye
| | - Merve Beker
- Department of Medical Biology, International School of Medicine, University of Health Sciences Türkiye, Mekteb-i Tıbbiye-i Şahane (Hamidiye) Külliyesi Selimiye Mah., Tıbbiye Cad. No: 38, Üsküdar, 34668, Istanbul, Türkiye.
| |
Collapse
|
31
|
Wang Z, Li Z, Yang R, Wang Y, Ma W, Lv X, Gao J, Li Y. Rubidium ions as a novel therapeutic approach for glioblastoma. Sci Rep 2025; 15:12917. [PMID: 40234687 PMCID: PMC12000443 DOI: 10.1038/s41598-025-97688-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 04/07/2025] [Indexed: 04/17/2025] Open
Abstract
Glioblastoma (GBM) is a highly aggressive brain tumor with limited treatment options, mainly due to challenges such as incomplete resection and blood-brain barrier limitations. Rubidium, a naturally occurring alkali metal with favorable biocompatibility, widely used in myocardial and tumor perfusion imaging as a blood flow tracer, is repurposed in this study to investigate its potential therapeutic effects and mechanisms against GBM. The impacts of rubidium ions (Rb⁺) on GBM cells were assessed through functional assays evaluating proliferation, migration, invasion, and apoptosis. RNA sequencing and Western blot analyses were employed to investigate molecular mechanisms, while in vivo models were used to evaluate therapeutic efficacy and safety. Rb⁺ treatment significantly suppressed GBM cell proliferation, migration, and invasion, while inducing apoptosis and cell cycle arrest at the G2/M phase. Mechanistic studies revealed that Rb⁺ downregulated the phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) pathway, contributing to the induction of apoptosis in tumor cells. In vivo, Rb⁺ exhibited potent anti-tumor activity with no detectable adverse effects on major organs, physiological functions, or behavior in mice. Our findings highlight Rb⁺ as a promising and innovative candidate for GBM therapy, leveraging the PI3K/AKT/mTOR pathway to inhibit tumor growth and promote apoptosis. This study underscores the potential of Rb⁺ in addressing the urgent need for novel GBM treatments, warranting further preclinical and clinical investigations.
Collapse
Affiliation(s)
- Zairan Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, 100000, China
| | - Zhimin Li
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, 100000, China
| | - Ran Yang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100000, China
| | - Yu Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, 100000, China
| | - Wenbin Ma
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, 100000, China
| | - Xiang Lv
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100000, China
| | - Jun Gao
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, 100000, China.
| | - Yongning Li
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, 100000, China.
| |
Collapse
|
32
|
Wu J, Qiu Y, Tian M, Wang L, Gao K, Yang X, Jiang Z. Flavonoids from Scutellaria baicalensis: Promising Alternatives for Enhancing Swine Production and Health. Int J Mol Sci 2025; 26:3703. [PMID: 40332337 PMCID: PMC12027786 DOI: 10.3390/ijms26083703] [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: 03/05/2025] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 05/08/2025] Open
Abstract
Concerns over vaccine safety, bacterial resistance, and drug residues have led to increased interest in plant extracts for improving swine nutrition and health. Scutellaria baicalensis Georgi, rich in four primary flavonoids-baicalin, baicalein, wogonoside, and wogonin-demonstrates significant pharmacological properties, including anti-inflammatory, antioxidant, antibacterial, and antiviral activities in swine. These flavonoids have been shown to enhance growth performance, improve immunity, modulate gut microbiota, and aid in the prevention and treatment of various diseases. This review highlights the pharmacological effects of these flavonoids in swine, with a focus on network pharmacology to reveal the underlying molecular mechanisms. By constructing drug-target networks and identifying key signaling pathways, the review reveals how these flavonoids interact with biological systems to promote health. Furthermore, it discusses the practical applications of Scutellaria baicalensis flavonoids in swine production and outlines potential future research directions. This work provides a theoretical framework for understanding the therapeutic targets of these flavonoids, offering valuable insights for advancing sustainable and healthy pig farming practices.
Collapse
Affiliation(s)
- Jing Wu
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (J.W.); (Y.Q.); (M.T.); (L.W.); (K.G.); (Z.J.)
- State Key Laboratory of Swine and Poultry husbandry Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Key Laboratory of Animal Husbandry and Nutrition, Guangzhou 510640, China
| | - Yueqin Qiu
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (J.W.); (Y.Q.); (M.T.); (L.W.); (K.G.); (Z.J.)
- State Key Laboratory of Swine and Poultry husbandry Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Key Laboratory of Animal Husbandry and Nutrition, Guangzhou 510640, China
| | - Min Tian
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (J.W.); (Y.Q.); (M.T.); (L.W.); (K.G.); (Z.J.)
- State Key Laboratory of Swine and Poultry husbandry Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Key Laboratory of Animal Husbandry and Nutrition, Guangzhou 510640, China
| | - Li Wang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (J.W.); (Y.Q.); (M.T.); (L.W.); (K.G.); (Z.J.)
- State Key Laboratory of Swine and Poultry husbandry Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Key Laboratory of Animal Husbandry and Nutrition, Guangzhou 510640, China
| | - Kaiguo Gao
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (J.W.); (Y.Q.); (M.T.); (L.W.); (K.G.); (Z.J.)
- State Key Laboratory of Swine and Poultry husbandry Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Key Laboratory of Animal Husbandry and Nutrition, Guangzhou 510640, China
| | - Xuefen Yang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (J.W.); (Y.Q.); (M.T.); (L.W.); (K.G.); (Z.J.)
- State Key Laboratory of Swine and Poultry husbandry Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Key Laboratory of Animal Husbandry and Nutrition, Guangzhou 510640, China
| | - Zongyong Jiang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China; (J.W.); (Y.Q.); (M.T.); (L.W.); (K.G.); (Z.J.)
- State Key Laboratory of Swine and Poultry husbandry Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Key Laboratory of Animal Husbandry and Nutrition, Guangzhou 510640, China
| |
Collapse
|
33
|
Querfurth HW, Lemere C, Ciola J, Havas D, Xia W, Lee HK. Target Validation Studies of PS48, a PDK-1 Allosteric Agonist, for the Treatment of Alzheimer's Disease Phenotype in APP/PS1 Transgenic Mice. Int J Mol Sci 2025; 26:3473. [PMID: 40331945 PMCID: PMC12027031 DOI: 10.3390/ijms26083473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/28/2025] [Accepted: 03/31/2025] [Indexed: 05/08/2025] Open
Abstract
The Alzheimer's disease (AD)-affected brain is known to be deficient in the utilization of glucose, its main energy substrate, and systemic diabetes is a significant risk factor for AD. In the course of biochemical and molecular investigations into this puzzling relationship, it has been shown that resistance to insulin action is a prominent feature of early stages of AD in the brain, thereby contributing to an energy failure state and a decline in synaptic function. In one AD-like cellular model, we found that β-amyloid (Aβ) accumulation inhibited insulin signaling and cell viability through an alteration of the PI3K/PDK-1/Akt signal pathway, an effect overcome by mTORC2 stimulation. A PDK-1 allosteric agonist, PS48, as well as newly synthesized analogs, were also found to reverse the metabolic defects caused by intracellular Aβ42 accumulation. In vivo, we previously showed that oral dosing of PS48 significantly improves learning and memory in APP/PS1 transgenic mice. Herein, we present evidence using unbiased immunohistological quantification and Western blot analyses demonstrating that ingested PS48 crosses into brain tissue where it targeted Akt and GSK3-β activities. Beneficial effects on neuronal number and Tau phosphorylation were found. Not unexpectedly, Aβ levels remained unchanged. These results support a path toward a future therapeutic trial of this untested strategy and agent in humans.
Collapse
Affiliation(s)
- Henry W. Querfurth
- Tufts Medical Center, Department of Neurology and Tufts University School of Medicine, Department of Neuroscience, 800 Washington St., and 136 Harrison Ave., Boston, MA 02111, USA
| | - Cynthia Lemere
- Brigham and Women’s Hospital, ARCND, 60 Fenwood Rd., Hale Bldg. for Transformative Medicine, Boston, MA 02115, USA; (C.L.); (J.C.)
| | - Jason Ciola
- Brigham and Women’s Hospital, ARCND, 60 Fenwood Rd., Hale Bldg. for Transformative Medicine, Boston, MA 02115, USA; (C.L.); (J.C.)
| | - Daniel Havas
- Psychogenics Inc., 215 College Rd., Paramus, NJ 07652, USA;
| | - Weiming Xia
- Chobanian and Avedisian School of Medicine, Department of Pharmacology, Physiology and Biophysics, Boston University, 72 E. Concord St., Boston, MA 02118, USA;
| | - Han Kyu Lee
- Tufts Medical Center, Department of Neurology, 800 Washington St., Boston, MA 02111, USA;
| |
Collapse
|
34
|
Antonyan L, Zhang X, Ni A, Peng H, Alsuwaidi S, Fleming P, Zhang Y, Semenak A, Macintosh J, Wu H, Hettige NC, Jefri M, Ernst C. Reciprocal and non-reciprocal effects of clinically relevant SETBP1 protein dosage changes. Hum Mol Genet 2025; 34:651-667. [PMID: 39825586 PMCID: PMC11973901 DOI: 10.1093/hmg/ddaf003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 12/17/2024] [Accepted: 01/06/2025] [Indexed: 01/20/2025] Open
Abstract
Many genes in the human genome encode proteins that are dosage sensitive, meaning they require protein levels within a narrow range to properly execute function. To investigate if clinically relevant variation in protein levels impacts the same downstream pathways in human disease, we generated cell models of two SETBP1 syndromes: Schinzel-Giedion Syndrome (SGS) and SETBP1 haploinsufficiency disease (SHD), where SGS is caused by too much protein, and SHD is caused by not enough SETBP1. Using patient and sex-matched healthy first-degree relatives from both SGS and SHD SETBP1 cases, we assessed how SETBP1 protein dosage affects downstream pathways in human forebrain progenitor cells. We find that extremes of SETBP1 protein dose reciprocally influence important signalling molecules such as AKT, suggesting that the SETBP1 protein operates within a narrow dosage range and that extreme doses are detrimental. We identified SETBP1 nuclear bodies as interacting with the nuclear lamina and suggest that SETBP1 may organize higher order chromatin structure via links to the nuclear envelope. SETBP1 protein doses may exert significant influence on global gene expression patterns via these SETBP1 nuclear bodies. This work provides evidence for the importance of SETBP1 protein dose in human brain development, with implications for two neurodevelopmental disorders.
Collapse
Affiliation(s)
- Lilit Antonyan
- Department of Human Genetics, McGill University, 3666 McTavish Street, Montreal, QC H3A 1Y2, Canada
- Rare Neurodevelopmental Disorders Laboratory, Montreal Neurological Institute, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Xin Zhang
- Rare Neurodevelopmental Disorders Laboratory, Montreal Neurological Institute, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Anjie Ni
- Department of Human Genetics, McGill University, 3666 McTavish Street, Montreal, QC H3A 1Y2, Canada
- Rare Neurodevelopmental Disorders Laboratory, Montreal Neurological Institute, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Huashan Peng
- Rare Neurodevelopmental Disorders Laboratory, Montreal Neurological Institute, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Shaima Alsuwaidi
- Rare Neurodevelopmental Disorders Laboratory, Montreal Neurological Institute, 3801 University Street, Montreal, QC H3A 2B4, Canada
- Integrated Program in Neuroscience, McGill University, 1033 Pine Ave. W., Montreal, QC H3A 1A1, Canada
| | - Peter Fleming
- Rare Neurodevelopmental Disorders Laboratory, Montreal Neurological Institute, 3801 University Street, Montreal, QC H3A 2B4, Canada
- Integrated Program in Neuroscience, McGill University, 1033 Pine Ave. W., Montreal, QC H3A 1A1, Canada
| | - Ying Zhang
- Rare Neurodevelopmental Disorders Laboratory, Montreal Neurological Institute, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Amelia Semenak
- Rare Neurodevelopmental Disorders Laboratory, Montreal Neurological Institute, 3801 University Street, Montreal, QC H3A 2B4, Canada
- Integrated Program in Neuroscience, McGill University, 1033 Pine Ave. W., Montreal, QC H3A 1A1, Canada
| | - Julia Macintosh
- Rare Neurodevelopmental Disorders Laboratory, Montreal Neurological Institute, 3801 University Street, Montreal, QC H3A 2B4, Canada
- Integrated Program in Neuroscience, McGill University, 1033 Pine Ave. W., Montreal, QC H3A 1A1, Canada
| | - Hanrong Wu
- Rare Neurodevelopmental Disorders Laboratory, Montreal Neurological Institute, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Nuwan C Hettige
- Department of Human Genetics, McGill University, 3666 McTavish Street, Montreal, QC H3A 1Y2, Canada
- Rare Neurodevelopmental Disorders Laboratory, Montreal Neurological Institute, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Malvin Jefri
- Rare Neurodevelopmental Disorders Laboratory, Montreal Neurological Institute, 3801 University Street, Montreal, QC H3A 2B4, Canada
- Integrated Program in Neuroscience, McGill University, 1033 Pine Ave. W., Montreal, QC H3A 1A1, Canada
| | - Carl Ernst
- Department of Human Genetics, McGill University, 3666 McTavish Street, Montreal, QC H3A 1Y2, Canada
- Rare Neurodevelopmental Disorders Laboratory, Montreal Neurological Institute, 3801 University Street, Montreal, QC H3A 2B4, Canada
- Integrated Program in Neuroscience, McGill University, 1033 Pine Ave. W., Montreal, QC H3A 1A1, Canada
| |
Collapse
|
35
|
Navarro-Traxler AJ, Ghisolfi L, Lien EC, Toker A. The glycosyltransferase ALG3 is an AKT substrate that regulates protein N-glycosylation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.01.646556. [PMID: 40236010 PMCID: PMC11996567 DOI: 10.1101/2025.04.01.646556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
The PI3K/AKT signaling pathway is frequently dysregulated in cancer and controls key cellular processes such as survival, proliferation, metabolism and growth. Protein glycosylation is essential for proper protein folding and is also often deregulated in cancer. Cancer cells depend on increased protein folding to sustain oncogene-driven proliferation rates. The N-glycosyltransferase asparagine-linked glycosylation 3 homolog (ALG3), a rate-limiting enzyme during glycan biosynthesis, catalyzes the addition of the first mannose to glycans in an alpha-1,3 linkage. Here we show that ALG3 is phosphorylated downstream of the PI3K/AKT pathway in both growth factor-stimulated cells and PI3K/AKT hyperactive cancer cells. AKT directly phosphorylates ALG3 in the amino terminal region at Ser11/Ser13. CRISPR/Cas9-mediated depletion of ALG3 leads to improper glycan formation and induction of endoplasmic reticulum stress, the unfolded protein response, and impaired cell proliferation. Phosphorylation of ALG3 at Ser11/Ser13 is required for glycosylation of cell surface receptors EGFR, HER3 and E-cadherin. These findings provide a direct link between PI3K/AKT signaling and protein glycosylation in cancer cells.
Collapse
|
36
|
Santos VR, Jerow LG, LaSarge CL. Behavioral analyses in rodent models of tuberous sclerosis complex. Epilepsy Behav 2025; 165:110313. [PMID: 39978075 DOI: 10.1016/j.yebeh.2025.110313] [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: 01/04/2025] [Accepted: 02/09/2025] [Indexed: 02/22/2025]
Abstract
Tuberous sclerosis complex (TSC) is typically associated with epilepsy, but patients also present with a myriad of comorbid neuropsychiatric disorders. TSC is caused by mutations in the tuberous sclerosis complex genes 1 or 2 (TSC1, TSC2). This TSC1/2 complex serves as a negative regulator of the mammalian target of rapamycin (mTOR) signaling pathway, which plays a crucial role in regulating neuronal function, including cell proliferation, survival, growth, and protein synthesis. Mutations result in hyperactivation of the pathway. Animal models with mutations in Tsc1 or Tsc2 consistently exhibit epilepsy and behavioral phenotypes. Additionally, abnormal neuronal populations can impact the broader network, leading to deficits in learning and memory, anxiety-like behaviors, deficits in social behaviors, and perseverative and repetitive behaviors. This review aims to synthesize the existing animal literature linking TSC models to epileptogenesis and behavioral impairments, with insights on how modifications in TSC signaling influence both the structure and function of neurons and behavior. Understanding these relationships may provide valuable insights into potential therapeutic targets for managing epilepsy and neuropsychiatric disorders associated with TSC dysregulation.
Collapse
Affiliation(s)
- Victor Rodrigues Santos
- Department of Morphology, Biology Cell Graduate Program, Neuroscience Graduate Program, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| | - Lilian G Jerow
- Neuroscience Graduate Program, University of Cincinnati, OH, USA.
| | - Candi L LaSarge
- Neuroscience Graduate Program, University of Cincinnati, OH, USA; Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Anesthesiology, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
37
|
Sabovic I, De Toni L, Di Nisio A, Radu CM, Gabbia D, De Martin S, Ferlin A. Additive effect of Bisphenol A and Pefluoro-sulphoctanoic acid exposure at subacute toxic levels, on a murine model of sertoli cell. J Endocrinol Invest 2025; 48:951-958. [PMID: 39556264 DOI: 10.1007/s40618-024-02498-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 11/01/2024] [Indexed: 11/19/2024]
Abstract
PURPOSE Endocrine disruptors (EDs) interfere with the endocrine system leading to health consquences and reproductive derangements. Most EDs are environmental pollutants whose risk evaluation is hampered by the simultaneous exposure to a number of chemicals. Here we investigated the possible mechanistic involvement of Sertoli cells, the nurse cell population in the seminiferous tubule, in the reproductive toxicity of Bisphenol A (BPA) and perfluoro-octane sulphonate (PFOS), two acknowledged EDs, at recognized subacute toxic levels. METHODS Mouse Sertoli cell line TM4 were exposed for 24 h to 40 ng/mL BPA or 30 ng/mL PFOS or their association. Cell proliferation was measuerd by MTT assay. Cell apoptosis was evaluated with Annexin-V/propidium iodide staining. Protein expression analysis was peformed by western blotting. RESULTS Compared to unexposed controls (100.0 ± 3.5%), cells exposed to BPA (79.5 ± 3.5%) or PFOS (76.0 ± 7.9%) showed reduced survival rate (P < 0.001 vs control). The exposure to the mixture of BPA and PFOS was associated with a further reduction of cell survival (63.9 ± 7.2%, P < 0.001 vs control) and an increase of the percentage of apoptotic cells (13.7 ± 4.6% control, 40.3 ± 13.5% BPA, PFOS 28.7 ± 5.6%, 67.1 ± 19.6% BPA + PFOS P = 0.001 vs control; P = 0.022 vs BPA). The exposure to the combination of BPA and PFOS was associated with Akt-signaling pathway activation and with the downstream caspase 3 cleavage. In addition, the exposure to the combination of BPA and PFOS was associated with NF-κB activation and increased expression of FasL. CONCLUSION Subacute toxic levels of BPA and PFOS display additive effects on Sertoli cell apoptosis with the possible involvement of FasL-dependent germ cell apoptosis.
Collapse
Affiliation(s)
- I Sabovic
- Department of Medicine, Unit of Andrology and Reproductive Medicine, University of Padova, Padova, Italy
| | - L De Toni
- Department of Medicine, Unit of Andrology and Reproductive Medicine, University of Padova, Padova, Italy
| | - A Di Nisio
- Department of Medicine, Unit of Andrology and Reproductive Medicine, University of Padova, Padova, Italy
- Department of Health, Nutrition and Sport, Pegaso Telematic University, Naples, Italy
| | - C M Radu
- Department of Medicine, Thrombotic and Haemorrhagic Disease Unit and Haemophilia Center, University of Padova, Padova, Italy
| | - D Gabbia
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - S De Martin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - A Ferlin
- Department of Medicine, Unit of Andrology and Reproductive Medicine, University of Padova, Padova, Italy.
| |
Collapse
|
38
|
Akande AO, Carter ZA, Stokes KY, Nam HW. Endothelial Neurogranin Regulates Blood-Brain Barrier Permeability via Modulation of the AKT Pathway. Mol Neurobiol 2025; 62:3991-4007. [PMID: 39367201 PMCID: PMC11880131 DOI: 10.1007/s12035-024-04522-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 09/23/2024] [Indexed: 10/06/2024]
Abstract
Neurogranin (Ng) expression is a biomarker for Alzheimer's disease. A loss of brain Ng and an increase in CSF Ng positively correlate with cognitive decline. Ng is known to regulate neuronal calcium-calmodulin binding and synaptic plasticity, which are critical for learning/memory. Interestingly, we discovered that Ng is also expressed in mouse and human blood-brain barrier (BBB). However, the role of Ng expression in brain vasculature remains largely undefined. In this study, we investigated the role of Ng expression on neurovascular structure and function using Ng null mice and human cerebral microvascular endothelial (hCMEC/D3) cells. We performed brain clearing and immunolabeling of blood vessels from whole brains and brain slices. Deletion of Ng significantly decreases neurovascular density in mice. Using in vivo permeability assays, we found increased neurovascular permeability in Ng null mice. We also observed significant changes in the expression of tight junction proteins using western blot and immunofluorescent staining. To identify the molecular pathways involved, we carried out label-free proteomics on brain lysates from endothelial-specific Ng knockout mice. Ingenuity Pathway Analysis indicated that the AKT pathway is attenuated in the vasculature of endothelial-specific Ng knockout mice. To validate these in vivo findings, we pharmacologically manipulated AKT signaling in hCMEC/D3 cells and observed that inhibition of AKT activation causes increased permeability. Our results indicate that the loss of Ng expression alters neurovascular structure and permeability, potentially contributing to neurological dysfunction. Therefore, modulating Ng expression in the BBB may offer a novel therapeutic approach for Alzheimer's disease.
Collapse
Affiliation(s)
- Adesewa O Akande
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA, 71103, USA
| | - Zachary A Carter
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA, 71103, USA
| | - Karen Y Stokes
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, 71103, USA
| | - Hyung W Nam
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA, 71103, USA.
| |
Collapse
|
39
|
Theocharopoulos C, Ziogas IA, Mungo B, Gogas H, Ziogas DC, Kontis E. HER2-targeted therapies: Unraveling their role in biliary tract cancers. Crit Rev Oncol Hematol 2025; 208:104655. [PMID: 39923923 DOI: 10.1016/j.critrevonc.2025.104655] [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: 01/31/2025] [Accepted: 02/05/2025] [Indexed: 02/11/2025] Open
Abstract
Biliary tract cancers (BTCs) constitute a heterogeneous group of malignancies with rising incidence and limited therapeutic options in advanced stages, leading to increased overall mortality. Extensive genomic profiling has identified key oncogenic drivers in BTCs that represent promising therapeutic targets and could change the treatment paradigm. Evidence suggests improved survival outcomes for patients with actionable molecular alterations who received matched targeted therapies. Human epidermal growth factor receptor 2 (HER2) is a receptor tyrosine kinase and proto-oncogene that has been extensively studied as a prognostic biomarker and a therapeutic target in multiple solid organ malignancies. Recent clinical trials on the combination of trastuzumab with tucatinib, FOLFOX, or pertuzumab for previously treated, HER2-positive, advanced BTCs have shown improved outcomes compared to current second-line therapies. Early evidence from observational studies on trastuzumab-containing regimens as first-line suggests promising efficacy. Furthermore, the recent tumor-agnostic approval of trastuzumab deruxtecan for HER2-positive solid tumors has formally introduced HER2-directed agents in the BTC therapeutic arsenal. This review aims to summarize the rapidly evolving landscape of HER2-directed agents for BTCs, highlighting current evidence of survival benefit. Beginning with a concise presentation of the structural and functional aspects of HER2, we detail the frequency and prognostic significance of HER2 alterations in BTCs and discuss all available preclinical and clinical data on anti-HER2 agents tested for BTCs.
Collapse
Affiliation(s)
| | - Ioannis A Ziogas
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| | - Benedetto Mungo
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| | - Helen Gogas
- First Department of Internal Medicine, Laikon General Hospital, School of Medicine, National Kapodistrian University of Athens, Athens 11527, Greece.
| | - Dimitrios C Ziogas
- First Department of Internal Medicine, Laikon General Hospital, School of Medicine, National Kapodistrian University of Athens, Athens 11527, Greece.
| | - Elissaios Kontis
- Department of Surgery, Metaxa Cancer Hospital, Piraeus 18537, Greece.
| |
Collapse
|
40
|
Ding Z, Shao G, Li M. Targeting autophagy in premature ovarian failure: Therapeutic strategies from molecular pathways to clinical applications. Life Sci 2025; 366-367:123473. [PMID: 39971127 DOI: 10.1016/j.lfs.2025.123473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 01/31/2025] [Accepted: 02/14/2025] [Indexed: 02/21/2025]
Abstract
Premature ovarian failure (POF) is a condition where the ovaries lose their function before the age of 40, leading to significant impacts on reproductive health and overall well-being. Current treatment options are limited and often ineffective at restoring ovarian function. This review explores the role of autophagy- a cellular process that helps maintain homeostasis by recycling damaged components-in the development and potential treatment of POF. Autophagy is crucial for the survival of follicle cells and can be disrupted by various stressors associated with POF, such as oxidative damage and mitochondrial dysfunction. We review several key molecular pathways involved in autophagy, including the PI3K/AKT/mTOR, PINK1-Parkin, JAK2/STAT3, MAPK and AMPK/FOXO3a pathways, which have been implicated in POF. Each pathway offers unique insights into how autophagy can be modulated to counteract POF-related damage. Additionally, we discuss emerging therapeutic strategies that target these pathways, including chemical compounds, peptides, hormones, RNA therapy, extracellular vesicles and traditional Chinese medicine. These approaches aim to restore autophagic balance, promote follicle survival and improve ovarian function. By targeting autophagy, new treatments may offer hope for better management and potential reversal of POF, thus improving the quality of life for affected individuals.
Collapse
Affiliation(s)
- Ziwen Ding
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Genbao Shao
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Mingyang Li
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China.
| |
Collapse
|
41
|
Yao M, Zhu X, Chen YC, Yang GH, Ao P. Exploring Multi-Target Therapeutic Strategies for Glioblastoma via Endogenous Network Modeling. Int J Mol Sci 2025; 26:3283. [PMID: 40244148 PMCID: PMC11989339 DOI: 10.3390/ijms26073283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/25/2025] [Accepted: 03/29/2025] [Indexed: 04/18/2025] Open
Abstract
Medical treatment of glioblastoma presents a significant challenge. A conventional medication has limited effectiveness, and a single-target therapy is usually effective only in the early stage of the treatment. Recently, there has been increasing focus on multi-target therapies, but the vast range of possible combinations makes clinical experimentation and implementation difficult. From the perspective of systems biology, this study conducted simulations for multi-target glioblastoma therapy based on dynamic analysis of previously established endogenous networks, validated with glioblastoma single-cell RNA sequencing data. Several potentially effective target combinations were identified. The findings also highlight the necessity of multi-target rather than single-target intervention strategies in cancer treatment, as well as the promise in clinical applications and personalized therapies.
Collapse
Affiliation(s)
- Mengchao Yao
- Shanghai Center for Quantitative Life Sciences and Physics Department, Shanghai University, Shanghai 200444, China;
| | - Xiaomei Zhu
- Shanghai Key Laboratory of Modern Optical System, School of Optical-Electrical and Computer Engineering, University of Shanghai for Science and Technology, Shanghai 200444, China
| | - Yong-Cong Chen
- Shanghai Center for Quantitative Life Sciences and Physics Department, Shanghai University, Shanghai 200444, China;
| | - Guo-Hong Yang
- Department of Physics, Shanghai University, Shanghai 200444, China;
| | - Ping Ao
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| |
Collapse
|
42
|
Huang B, Wang H, An Z, Yang Z, Cao J, Wang L, Luo X, Qi H. Compromised Peroxisome Proliferator-Activated Receptor γ-Mediated Impaired Placental Glucose Transport Via the Phosphatidylinositol 3-Kinase/Protein Kinase B Signaling Pathway Is Associated With Fetal Growth Restriction. J Transl Med 2025; 105:104103. [PMID: 39909142 DOI: 10.1016/j.labinv.2025.104103] [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: 06/12/2024] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 02/07/2025] Open
Abstract
Fetal growth restriction (FGR) is a condition in which a fetus cannot grow to its full potential during pregnancy. It is a leading cause of perinatal mortality and morbidity. However, the underlying etiology remains elusive. Here, we report that peroxisome proliferator-activated receptor γ (PPARγ) is inactivated in the trophoblasts of the human placenta of FGR-complicated pregnancies. In the FGR placentas, p-PI3KTyr458 and p-AKTSer473 levels were also lowered. Additionally, there was a reduction in GLUT3 and GLUT4 levels in the cell membrane. Consistently, FGR patients showed decreased glucose concentrations in both the placenta and umbilical cord blood compared with that in normal pregnancy. In mouse models, deletion of Pparg in trophoblasts and reduced uterine perfusion pressure surgery successfully induced FGR and replicated these changes. Modulating PPARγ activity using rosiglitazone or GW9662 in BeWo cells, a model of syncytiotrophoblasts, resulted in the activation or inhibition of the phosphatidylinositol 3-kinase/protein kinase B signaling pathway, as well as the promotion or reduction of membrane translocation of GLUT3 and GLUT4, ultimately affecting glucose uptake in trophoblast cells. MK-2206 blocked these regulatory effects of rosiglitazone in BeWo cells. Furthermore, the administration of rosiglitazone encapsulated in placenta-targeting nanoparticles improved the growth and development of fetal mice in the reduced uterine perfusion pressure group. In summary, PPARγ in trophoblast cells orchestrates the translocation of GLUT3 and GLUT4 to the cellular membrane via the phosphatidylinositol 3-kinase/protein kinase B signaling pathway, thereby regulating cellular glucose uptake and transport. Dysfunctions in this mechanism are strongly associated with FGR. Therefore, targeted activation of PPARγ in the placenta may be a potentially efficacious intrauterine intervention for FGR.
Collapse
Affiliation(s)
- Biao Huang
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing, China; State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Ministry of Education International Collaborative Laboratory of Reproduction and Development, Chongqing Medical University, Chongqing, China
| | - Hao Wang
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Zhongling An
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Ministry of Education International Collaborative Laboratory of Reproduction and Development, Chongqing Medical University, Chongqing, China; Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhongmei Yang
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Ministry of Education International Collaborative Laboratory of Reproduction and Development, Chongqing Medical University, Chongqing, China; Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; The Department of Obstetrics and Gynecology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Jinfeng Cao
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Ministry of Education International Collaborative Laboratory of Reproduction and Development, Chongqing Medical University, Chongqing, China; Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lan Wang
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaofang Luo
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Ministry of Education International Collaborative Laboratory of Reproduction and Development, Chongqing Medical University, Chongqing, China; Reproductive Medicine Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Hongbo Qi
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Ministry of Education International Collaborative Laboratory of Reproduction and Development, Chongqing Medical University, Chongqing, China; Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
43
|
Barone M, Baccaro P, Molfino A. An Overview of Sarcopenia: Focusing on Nutritional Treatment Approaches. Nutrients 2025; 17:1237. [PMID: 40218995 PMCID: PMC11990658 DOI: 10.3390/nu17071237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2025] [Revised: 03/28/2025] [Accepted: 03/30/2025] [Indexed: 04/14/2025] Open
Abstract
Sarcopenia is a syndrome characterized by the progressive and generalized loss of skeletal muscle mass and strength. This condition is associated with physical disability, decreased quality of life, and increased mortality. Therefore, reducing the prevalence of sarcopenia could significantly lower healthcare costs. Sarcopenia can be classified into primary and secondary sarcopenia. The former is related to aging and begins after the fourth decade of life; after that, there is a muscle loss of around 8% per decade until age 70 years, which subsequently increases to 15% per decade. On the other hand, secondary sarcopenia can affect all individuals and may result from various factors including physical inactivity, malnutrition, endocrine disorders, neurodegenerative diseases, inflammation, and cachexia. Understanding the multiple mechanisms involved in the onset and progression of sarcopenia allows for us to develop strategies that can prevent, treat, or at least mitigate muscle loss caused by increased protein breakdown. One potential treatment of sarcopenia is based on nutritional interventions, including adequate caloric and protein intake and specific nutrients that support muscle health. Such nutrients include natural food rich in whey protein and omega-3 fatty acids as well as nutritional supplements like branched-chain amino acids, β-hydroxy-β-methylbutyrate, and vitamin D along with food for special medical purposes. It is important to emphasize that physical exercises, especially resistance training, not only promote muscle protein synthesis on their own but also work synergistically with nutritional strategies to enhance their effectiveness.
Collapse
Affiliation(s)
- Michele Barone
- Gastroenterology Unit, Department of Precision and Regenerative Medicine, University of Bari, Policlinic University Hospital, Piazza G. Cesare 11, 70124 Bari, Italy;
| | - Palmina Baccaro
- Gastroenterology Unit, Department of Precision and Regenerative Medicine, University of Bari, Policlinic University Hospital, Piazza G. Cesare 11, 70124 Bari, Italy;
| | - Alessio Molfino
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00185 Rome, Italy;
| |
Collapse
|
44
|
Xiang W, Yuan W, Ren L, Huang W, Liang H, Huang J, Luan L, Xu C, Hou Y. A case of quadruple wild-type gastrointestinal stromal tumor with CDC42BPB::NTRK3 fusion and abundant lymphoid infiltration. Diagn Pathol 2025; 20:31. [PMID: 40133893 PMCID: PMC11934696 DOI: 10.1186/s13000-025-01630-5] [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: 01/15/2025] [Accepted: 03/12/2025] [Indexed: 03/27/2025] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal tumors of the gastrointestinal tract. The most common mutations in GISTs are those in receptor tyrosine kinase (KIT) and platelet-derived growth factor receptor alpha (PDGFRA). GISTs without KIT or PDGFRA mutations are defined as wild-type (WT) GISTs. The molecular changes, prognosis, and treatments of WT GISTs remain uncertain. Among WT GISTs, neurotrophic tyrosine receptor kinase (NTRK) fusions have rarely been reported. We report a case of quadruple wild-type GIST harboring a novel CDC42BPB::NTRK3 fusion. In this study, we described a 66-year-old male patient with intrajejunal lesion. This case showed massive lymphocytic and plasma cell infiltration, which caused diagnostic difficulties in morphology. CDC42BPB::NTRK3 fusion was detected via next-generation sequencing (NGS), and this finding was confirmed by fluorescence in situ hybridization (FISH), which revealed NTRK3 breakage. However, the expression of the Trk protein in tumor tissue was not detected by immunohistochemistry (IHC). This finding expands the genetic spectrum of NTRK rearrangements in GISTs.
Collapse
Affiliation(s)
- Wentao Xiang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wei Yuan
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Lei Ren
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wen Huang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Huaiyu Liang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jie Huang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Lijuan Luan
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Chen Xu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yingyong Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
45
|
Kamel WA, Krishnaraj J, Ohki R. The Role of PHLDA3 in Cancer Progression and Its Potential as a Therapeutic Target. Cancers (Basel) 2025; 17:1069. [PMID: 40227573 PMCID: PMC11988131 DOI: 10.3390/cancers17071069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/12/2025] [Accepted: 03/18/2025] [Indexed: 04/15/2025] Open
Abstract
Pleckstrin homology-like domain family A, member 3 (PHLDA3) is a p53-regulated tumor suppressor protein that suppresses AKT-mediated survival and oncogenic signaling. The PHLDA3 gene has garnered significant attention due to its multifaceted roles in tumorigenesis, metastasis, and invasion. This review explores the complex interactions between PHLDA3 and key cellular processes involved in cancer, emphasizing its regulatory mechanisms and clinical relevance. PHLDA3 has been found to be a critical regulator of metastatic pathways, particularly through its influence on the epithelial-mesenchymal transition (EMT) and in cellular invasion. Its interactions with pivotal signaling pathways, such as the Phosphoinositide 3-kinases/Protein kinase B (PI3K/AKT), p53, and Wnt/β-catenin pathways, highlight its multifunctional roles in various cancer types. Additionally, we discuss the potential of PHLDA3 as both a prognostic biomarker and a therapeutic target, offering new insights into its potential in treating advanced-stage malignancies. This review provides a detailed analysis of the role of PHLDA3 in cancer progression, including metastasis and invasion, underscoring its therapeutic potential.
Collapse
Affiliation(s)
- Walied A. Kamel
- Laboratory of Fundamental Oncology, National Cancer Center Research Institute, Tsukiji 5-1-1, Chuo-ku, Tokyo 104-0045, Japan; (W.A.K.); (J.K.)
- Department of Zoology, School of Science, Mansoura University, Mansoura 35516, Egypt
| | - Jayaraman Krishnaraj
- Laboratory of Fundamental Oncology, National Cancer Center Research Institute, Tsukiji 5-1-1, Chuo-ku, Tokyo 104-0045, Japan; (W.A.K.); (J.K.)
| | - Rieko Ohki
- Laboratory of Fundamental Oncology, National Cancer Center Research Institute, Tsukiji 5-1-1, Chuo-ku, Tokyo 104-0045, Japan; (W.A.K.); (J.K.)
| |
Collapse
|
46
|
Shi H, Yang Y, Gao J, Kumar S, Xie H, Chen Z, Lyu J, Sihto H, Koljonen V, Vega-Rubin-de-Celis S, Vukojevic V, Farnebo F, Björnhagen V, Höög A, Juhlin CC, Lee L, Wickström M, Becker JC, Johnsen JI, Larsson C, Lui WO. Kit-mediated autophagy suppression driven by a viral oncoprotein emerges as a crucial survival mechanism in Merkel cell carcinoma. Autophagy 2025:1-21. [PMID: 40108758 DOI: 10.1080/15548627.2025.2477385] [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: 03/17/2024] [Revised: 01/20/2025] [Accepted: 01/31/2025] [Indexed: 03/22/2025] Open
Abstract
The KIT/c-KIT proto-oncogene is frequently over-expressed in Merkel cell carcinoma (MCC), an aggressive skin cancer commonly caused by Merkel cell polyomavirus (MCPyV). Here, we demonstrated that truncated MCPyV-encoded large T-antigen (LT) suppressed macroautophagy/autophagy by stabilizing and sequestering KIT in the paranuclear compartment via binding VPS39. KIT engaged with phosphorylated BECN1, thereby enhancing its association with BCL2 while diminishing its interaction with the PIK3C3 complex. This process ultimately resulted in the suppression of autophagy. Depletion of KIT triggered both autophagy and apoptosis, and decreased LT expression. Conversely, blocking autophagy in KIT-depleted cells restored LT levels and rescued apoptosis. Additionally, stimulating autophagy efficiently increased cell death and inhibited tumor growth of MCC xenografts in mice. These insights into the interplay between MCPyV LT and autophagy regulation reveal important mechanisms by which viral oncoproteins are essential for MCC cell viability. Thus, autophagy-inducing agents represent a therapeutic strategy in advanced MCPyV-associated MCC.Abbreviation: 3-MA, 3-methyladenine; AL, autolysosome; AP, autophagosome; Baf-A1, bafilomycin A1; BARA, β-α repeated autophagy specific domain; BH3, BCL2 homology 3 domain; CCD, coiled-coil domain; CHX, cycloheximide; Co-IP, co-immunoprecipitation; CQ, chloroquine; CTR, control; DAPI, 4',6-diamidino-2-phenylindole; EBSS, Earle's balanced salt solution; ECD, evolutionarily conserved domain; EEE, three-tyrosine phosphomimetic mutations Y229E Y233E Y352E; ER, endoplasmic reticulum; FFF, three-tyrosine non-phosphomimetic mutations; FFPE, formalin-fixed paraffin-embedded; FL, full-length; GIST, gastrointestinal stromal tumor; IB, immunoblotting; IHC, immunohistochemistry; KIT-HEK293, KIT stably expressing HEK293 cells; KRT20/CK20, keratin 20; LT, large T-antigen; LT339, MCPyV truncated LT antigen; LTco, codon-optimized MCPyV LT antigen; MCC, Merkel cell carcinoma; MCPyV-, MCPyV-negative; MCPyV, Merkel cell polyomavirus; MCPyV+, MCPyV-positive; PARP1, poly(ADP-ribose) polymerase 1; PCI, pan-caspase inhibitor; PI, propidium iodide; PtdIns3K, class III phosphatidylinositol 3-kinase; PtdIns3P, phosphatidylinositol-3-phosphate; RB1, RB transcriptional corepressor 1; RTKs, receptor tyrosine kinases; KITLG/SCF, KIT ligand; sT, small T-antigen; sTco, codon-optimized MCPyV sT antigen; T-B, Tat-BECN1; T-S, Tat-scrambled; TEM, transmission electron microscopy.
Collapse
Affiliation(s)
- Hao Shi
- Department of Oncology-Pathology, Karolinska Institutet; BioClinicum J6:20, Karolinska University Hospital, Solna, Sweden
| | - Yajie Yang
- Department of Oncology-Pathology, Karolinska Institutet; BioClinicum J6:20, Karolinska University Hospital, Solna, Sweden
| | - Jiwei Gao
- Department of Oncology-Pathology, Karolinska Institutet; BioClinicum J6:20, Karolinska University Hospital, Solna, Sweden
- The Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Satendra Kumar
- Department of Oncology-Pathology, Karolinska Institutet; BioClinicum J6:20, Karolinska University Hospital, Solna, Sweden
| | - Hong Xie
- Department of Oncology-Pathology, Karolinska Institutet; BioClinicum J6:20, Karolinska University Hospital, Solna, Sweden
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Ziqing Chen
- Department of Oncology-Pathology, Karolinska Institutet; BioClinicum J6:20, Karolinska University Hospital, Solna, Sweden
| | - Jiawen Lyu
- Department of Oncology-Pathology, Karolinska Institutet; BioClinicum J6:20, Karolinska University Hospital, Solna, Sweden
| | - Harri Sihto
- Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Virve Koljonen
- Department of Plastic Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | | | - Vladana Vukojevic
- Laboratory of Experimental Alcohol and Drug Addiction Research, Department of Clinical Neuroscience, Karolinska Institutet; Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Filip Farnebo
- Department of Molecular Medicine and Surgery, Karolinska Institutet; Clinic for Reconstructive Plastic Surgery, Karolinska University Hospital, Stockholm, Sweden
| | - Viveca Björnhagen
- Department of Molecular Medicine and Surgery, Karolinska Institutet; Clinic for Reconstructive Plastic Surgery, Karolinska University Hospital, Stockholm, Sweden
| | - Anders Höög
- Department of Oncology-Pathology, Karolinska Institutet; BioClinicum J6:20, Karolinska University Hospital, Solna, Sweden
- Department of Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden
| | - C Christofer Juhlin
- Department of Oncology-Pathology, Karolinska Institutet; BioClinicum J6:20, Karolinska University Hospital, Solna, Sweden
- Department of Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden
| | - Linkiat Lee
- Department of Oncology-Pathology, Karolinska Institutet; BioClinicum J6:20, Karolinska University Hospital, Solna, Sweden
| | - Malin Wickström
- Department of Women's and Children's Health, Karolinska Institutet; BioClinicum J5:20, Karolinska University Hospital, Solna, Sweden
| | - Jürgen C Becker
- Translational Skin Cancer Research, University Medicine Essen, Essen, Germany
- German Cancer Consortium (DKTK), German Cancer Research Institute (DKFZ), Heidelberg, Baden-Württemberg, Germany
| | - John Inge Johnsen
- Department of Women's and Children's Health, Karolinska Institutet; BioClinicum J5:20, Karolinska University Hospital, Solna, Sweden
| | - Catharina Larsson
- Department of Oncology-Pathology, Karolinska Institutet; BioClinicum J6:20, Karolinska University Hospital, Solna, Sweden
| | - Weng-Onn Lui
- Department of Oncology-Pathology, Karolinska Institutet; BioClinicum J6:20, Karolinska University Hospital, Solna, Sweden
| |
Collapse
|
47
|
Waheed I, Sikandri T, Zaheen S, Khakwani MMAK, An Z, Liu T, Zhu C, Wei J. Evaluating the Molecular Interactions between Type 2 Diabetes Mellitus and Parkinson's Disease: Role of Antidiabetic Drugs as Promising Therapeutics. ACS Chem Neurosci 2025; 16:988-999. [PMID: 40042145 DOI: 10.1021/acschemneuro.4c00819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025] Open
Abstract
Evidence from previous research demonstrates a relationship between diabetes mellitus (DM) and Parkinson's disease (PD). T2DM is associated with chronic glucose dysregulation, as an etiological factor. It inhibits neuronal function through disrupted insulin signaling and oxidative stress, which ultimately lead to the loss of dopaminergic neurons in the substantia nigra (SN). Interactions between T2DM and PD were analyzed by gene expression, coexpression, and gene set enrichment via NCBI and STRING databases following pathways like KEGG and Reactome. The study identified nine key gene interactions through published literature on different databases and search engines that are involved in the progression of these chronic diseases. Furthermore, some genetic and nongenetic risk factors, gene mutations and environmental factors, are also involved in the progression of T2DM and PD. This review highlights the limitations of currently available drug treatments for these diseases and examines modern therapeutic approaches to address neurodegenerative and metabolic abnormalities. We critically assess the current experimental methodologies aimed at unraveling the pathophysiological mechanisms linking PD and T2DM while addressing the key challenges impeding a comprehensive understanding of the concurrent emergence of these debilitating age-related conditions.
Collapse
Affiliation(s)
- Irum Waheed
- Institute for Brain Sciences Research, Center for Translational Neuromedicine and Neurourology, Huaihe Hospital of Henan University, School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Talal Sikandri
- Institute for Brain Sciences Research, Center for Translational Neuromedicine and Neurourology, Huaihe Hospital of Henan University, School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Sumbal Zaheen
- Institute for Brain Sciences Research, Center for Translational Neuromedicine and Neurourology, Huaihe Hospital of Henan University, School of Life Sciences, Henan University, Kaifeng 475004, China
| | | | - Zhaowu An
- Institute for Brain Sciences Research, Center for Translational Neuromedicine and Neurourology, Huaihe Hospital of Henan University, School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Tingting Liu
- Institute for Brain Sciences Research, Center for Translational Neuromedicine and Neurourology, Huaihe Hospital of Henan University, School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Chaoyang Zhu
- Institute for Brain Sciences Research, Center for Translational Neuromedicine and Neurourology, Huaihe Hospital of Henan University, School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Jianshe Wei
- Institute for Brain Sciences Research, Center for Translational Neuromedicine and Neurourology, Huaihe Hospital of Henan University, School of Life Sciences, Henan University, Kaifeng 475004, China
| |
Collapse
|
48
|
Mir MM, Jeelani M, Alharthi MH, Rizvi SF, Sohail SK, Wani JI, Sabah ZU, BinAfif WF, Nandi P, Alshahrani AM, Alfaifi J, Jehangir A, Mir R. Unraveling the Mystery of Insulin Resistance: From Principle Mechanistic Insights and Consequences to Therapeutic Interventions. Int J Mol Sci 2025; 26:2770. [PMID: 40141412 PMCID: PMC11942988 DOI: 10.3390/ijms26062770] [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: 02/15/2025] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 03/28/2025] Open
Abstract
Insulin resistance (IR) is a significant factor in the development and progression of metabolic-related diseases like dyslipidemia, T2DM, hypertension, nonalcoholic fatty liver disease, cardiovascular and cerebrovascular disorders, and cancer. The pathogenesis of IR depends on multiple factors, including age, genetic predisposition, obesity, oxidative stress, among others. Abnormalities in the insulin-signaling cascade lead to IR in the host, including insulin receptor abnormalities, internal environment disturbances, and metabolic alterations in the muscle, liver, and cellular organelles. The complex and multifaceted characteristics of insulin signaling and insulin resistance envisage their thorough and comprehensive understanding at the cellular and molecular level. Therapeutic strategies for IR include exercise, dietary interventions, and pharmacotherapy. However, there are still gaps to be addressed, and more precise biomarkers for associated chronic diseases and lifestyle interventions are needed. Understanding these pathways is essential for developing effective treatments for IR, reducing healthcare costs, and improving quality of patient life.
Collapse
Affiliation(s)
- Mohammad Muzaffar Mir
- Department of Clinical Biochemistry, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia
| | - Mohammed Jeelani
- Department of Physiology, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia;
| | - Muffarah Hamid Alharthi
- Department of Family and Community Medicine, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia; (M.H.A.); (P.N.)
| | - Syeda Fatima Rizvi
- Department of Pathology, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia; (S.F.R.); (S.K.S.)
| | - Shahzada Khalid Sohail
- Department of Pathology, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia; (S.F.R.); (S.K.S.)
| | - Javed Iqbal Wani
- Department of Internal Medicine, College of Medicine, King Khalid University, Abha 61421, Saudi Arabia; (J.I.W.); (Z.U.S.)
| | - Zia Ul Sabah
- Department of Internal Medicine, College of Medicine, King Khalid University, Abha 61421, Saudi Arabia; (J.I.W.); (Z.U.S.)
| | - Waad Fuad BinAfif
- Department of Internal Medicine, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia;
| | - Partha Nandi
- Department of Family and Community Medicine, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia; (M.H.A.); (P.N.)
| | - Abdullah M. Alshahrani
- Department of Family and Community Medicine, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia; (M.H.A.); (P.N.)
| | - Jaber Alfaifi
- Department of Child Health, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia;
| | - Adnan Jehangir
- Biomedical Sciences Department, College of Medicine, King Faisal University, Al Ahsa 31982, Saudi Arabia;
| | - Rashid Mir
- Prince Fahd Bin Sultan Research Chair, Department of MLT, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia;
| |
Collapse
|
49
|
Zhong T, Wu S, Chen G, Zhan S, Wang L, Cao J, Guo J, Li L, Zhang H, Niu L. Integrated analyses of transcriptomes, metabolomes, and proteomes unveil the role of FoXO signaling axis in buck semen cryopreservation. Theriogenology 2025; 235:19-30. [PMID: 39756112 DOI: 10.1016/j.theriogenology.2024.12.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 11/03/2024] [Accepted: 12/29/2024] [Indexed: 01/07/2025]
Abstract
Sperm cryopreservation is a complex process involving gene expression, protein synthesis, membrane stability, and metabolic adaptation. However, molecular alterations in sperm cryopreservation and the mechanisms defending against freezing damage remain poorly understood. This study investigates these changes and defense mechanisms using transcriptomics, proteomics, and metabolomics data. During sperm cryopreservation, the expression level of G protein subunit alpha i3 (GNAI3) was significantly downregulated in post-thaw sperm (P < 0.001), while matrix metallopeptidase 9 (MMP9) was upregulated compared to FS groups (P < 0.01). Additionally, interleukin 6 (IL6) expression in the CS group showed an approximate increase (P < 0.05), whereas ribosomal protein S27a (RPS27A) expression decreased markedly (P < 0.05). Other important molecules such as macrophage stimulating 1 receptor (MST1R), hypoxia-inducible factor 1 subunit alpha (HIF1A), fibroblast growth factor 8 (FGF8), CD9 molecule (CD9), peptidase D (PEPD) and terminal nucleotidyltransferase 5B (TENT5B) also exhibited significant changes in expression (P < 0.05). Moreover, the study revealed the regulatory roles of metabolites such as glucose and glutamic acid during sperm cryopreservation. The involvement of catalase (CAT) protein in antioxidant defense was also noted. The interactions among mRNAs, miRNAs, proteins, and metabolites highlight the critical role of the FoxO signaling pathway in modulating responses to freezing. Our study reveals the molecular regulatory mechanisms of sperm during cryopreservation, emphasizing the importance of the FoxO pathway and specific metabolites in response to cryo-injury. These findings provide deeper insights into the complexity of sperm cryobiology and offer practical guidance for optimizing sperm cryopreservation.
Collapse
Affiliation(s)
- Tao Zhong
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Shun Wu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
| | - Guolin Chen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
| | - Siyuan Zhan
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
| | - Linjie Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
| | - Jiaxue Cao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
| | - Jiazhong Guo
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
| | - Li Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
| | - Hongping Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
| | - Lili Niu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
| |
Collapse
|
50
|
Waheed YA, Buberwa W, Sun D. Glial cell line-derived neurotrophic factor and its role in attenuating renal fibrosis: a review. Korean J Intern Med 2025; 40:219-229. [PMID: 38086618 PMCID: PMC11938710 DOI: 10.3904/kjim.2023.246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/08/2023] [Accepted: 09/01/2023] [Indexed: 03/20/2025] Open
Abstract
Chronic kidney disease is estimated to affect approximately 10 to 15% of the Chinese population. Renal fibrosis is characterized by progressive extracellular matrix deposition in the kidney parenchyma with eventual tissue scarring and inevitable deterioration of renal function. Vascular rarefaction, glomerulosclerosis, interstitial inflammation, and fibrogenesis are associated with or contribute to renal fibrosis. Recent studies have revealed that glial cell-derived neurotrophic factor (GDNF) is involved in kidney morphogenesis and amelioration of renal injury. Ideal therapies targeting the pathogenesis of renal fibrosis should have the potential to inhibit glomerular and tubulointerstitial fibrosis by targeting multiple pathological events. GDNF plays a unique role in both renal development and improvement of renal fibrosis, and GDNF kidney receptors and signaling pathways can ameliorate renal apoptosis and inflammation. Our work contributes to the establishment of GDNF as an emerging therapy that can increase the effectiveness of currently used interventions to improve renal fibrosis. This literature review focuses on the important role of GDNF in renal development and its relationship with renal fibrosis.
Collapse
Affiliation(s)
| | - Wokuheleza Buberwa
- Department of Pediatrics, Arusha Lutheran Medical Center, Arusha,
Tanzania
| | - Dong Sun
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou,
China
- Department of Internal Medicine and Diagnostics, Xuzhou Medical University, Xuzhou,
China
| |
Collapse
|