1
|
Zou Z, Zhong L. Anaplastic thyroid cancer: Genetic roles, targeted therapy, and immunotherapy. Genes Dis 2025; 12:101403. [PMID: 40271195 PMCID: PMC12018003 DOI: 10.1016/j.gendis.2024.101403] [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: 02/24/2024] [Revised: 07/02/2024] [Accepted: 08/02/2024] [Indexed: 04/25/2025] Open
Abstract
Anaplastic thyroid cancer (ATC) stands as the most formidable form of thyroid malignancy, presenting a persistent challenge in clinical management. Recent years have witnessed a gradual unveiling of the intricate genetic underpinnings governing ATC through next-generation sequencing. The emergence of this genetic landscape has paved the way for the exploration of targeted therapies and immunotherapies in clinical trials. Despite these strides, the precise mechanisms governing ATC pathogenesis and the identification of efficacious treatments demand further investigation. Our comprehensive review stems from an extensive literature search focusing on the genetic implications, notably the pivotal MAPK and PI3K-AKT-mTOR signaling pathways, along with targeted therapies and immunotherapies in ATC. Moreover, we screen and summarize the advances and challenges in the current diagnostic approaches for ATC, including the invasive tissue sampling represented by fine needle aspiration and core needle biopsy, immunohistochemistry, and 18F-fluorodeoxyglucose positron emission tomography/computed tomography. We also investigate enormous studies on the prognosis of ATC and outline independent prognostic factors for future clinical assessment and therapy for ATC. By synthesizing this literature, we aim to encapsulate the evolving landscape of ATC oncology, potentially shedding light on novel pathogenic mechanisms and avenues for therapeutic exploration.
Collapse
Affiliation(s)
- Zhao Zou
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Linhong Zhong
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging and Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
2
|
Tufail M. PTEN-mediated resistance in cancer: From foundation to future therapies. Toxicol Rep 2025; 14:101987. [PMID: 40129883 PMCID: PMC11930710 DOI: 10.1016/j.toxrep.2025.101987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 03/01/2025] [Accepted: 03/02/2025] [Indexed: 03/26/2025] Open
Abstract
In cancer resistance, phosphatase and tensin homolog deleted (PTEN) has emerged as a prominent protagonist. PTEN exerts its influence by regulating crucial signaling pathways that govern cell proliferation, survival, and differentiation. This comprehensive review article investigates deeply into the complex realm of PTEN-mediated drug resistance mechanisms in cancers. Our journey begins by exploring PTEN's foundational role of PTEN, unveiling its significance as a molecular conductor that intricately coordinates vital cellular pathways. We thoroughly dissected the intricate milieu of PTEN alterations, including mutations, deletions, and epigenetic silencing, and elucidated their profound implications for fueling cancer growth and evading treatment. As we navigate the complex network of PTEN, we unravel the intricate interplay between PTEN and pivotal signaling pathways, such as PI3K/AKT, MAPK/ERK, and Wnt/β-catenin, further complicating the resistance landscape. This expedition, through these intricately intertwined signaling cascades, provides insight into the multifaceted mechanisms driving resistance, thereby revealing potential exploitable weaknesses. In our quest for therapeutic strategies, we need to explore innovative approaches to restore PTEN function, encompassing genetic therapies, pharmacological agents, and precision medicines tailored to PTEN status. The concept of combination therapy has emerged as a potent tool to overcome PTEN-associated resistance, offering promising synergistic interactions with standard treatments, targeted therapies, or immunotherapy. This review offers a comprehensive overview of PTEN-mediated drug resistance mechanisms in cancer and elucidates intricate interactions within this complex landscape. This underscores the central role of PTEN in drug resistance and provides valuable insights into promising strategies with the potential to reshape the future of cancer treatment.
Collapse
|
3
|
Kavitha L, Ranganathan K. Loss of Heterozygosity in Oral Potentially Malignant Disorders and Oral Squamous Cell Carcinoma - A Scoping Review. Head Neck Pathol 2025; 19:49. [PMID: 40278970 DOI: 10.1007/s12105-025-01787-x] [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: 02/19/2025] [Accepted: 04/01/2025] [Indexed: 04/26/2025]
Abstract
INTRODUCTION This scoping review was conducted to ascertain the loss of heterozygosity (LOH) signatures reported in Oral Potentially Malignant Disorders (OPMD) and Oral Squamous Cell Carcinoma (OSCC), in the literature in the last fifty years. METHODS The Joanna Briggs Institute recommendations (2023) for scoping review were used to extract, analyze, and present the results. The review was reported according to the PRISMA guidelines for Scoping Reviews (PRISMA-ScR). The most commonly reported genes associated with LOH in OPMD and OSCC are discussed. The Gene Ontology functional enrichment analysis gives the significance of the protein-protein interactions (PPI) of these genes using the STRING database. RESULTS An exhaustive database search of the title, abstract, and full-text screening consistent with the eligibility criteria yielded 277 studies. LOH commonly studied in OPMD and OSCC include p53 gene, p16 gene, adenomatous polyposis coli gene, retinoblastoma (Rb) gene, fragile histidine triad (FHIT) gene and phosphatase and tensin homolog (PTEN) gene. Chromosome loci involving 17p, 9p, 5q, 13q, 3p, and 10q were frequently reported in OPMD and OSCC. PPI analysis demonstrated strong evidence of p53 interaction with p16, FHIT, and Rb. CONCLUSION Distinctive signatures of LOH are seen in OPMD and OSCC. The LOH patterns identified in this scoping review underline the significance of advanced molecular techniques and the need for long-term prospective cohorts to understand LOH pathophysiology in oral carcinogenesis to enable their usefulness as biomarkers in early diagnosis, treatment, and prognostication of oral cancer.
Collapse
Affiliation(s)
- L Kavitha
- Department of Oral and Maxillofacial Pathology, Ragas Dental Colege and Hospital (Affiliated to The Tamil Nadu Dr. M.G.R. Medical University), ECR, Uthandi, Chennai, Tamil Nadu, 600 119, India
| | - K Ranganathan
- Department of Oral and Maxillofacial Pathology, Ragas Dental Colege and Hospital (Affiliated to The Tamil Nadu Dr. M.G.R. Medical University), ECR, Uthandi, Chennai, Tamil Nadu, 600 119, India.
| |
Collapse
|
4
|
Song Y, Ren X, Xiong J, Wang W, Zhao Q, Chang J, Yu B. Ubiquitin-Specific Protease 7 (USP7) as a Promising Therapeutic Target for Drug Discovery: From Mechanisms to Therapies. J Med Chem 2025; 68:7914-7931. [PMID: 40237780 DOI: 10.1021/acs.jmedchem.5c00102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
Protein ubiquitination is a reversible post-translational modification regulated by ubiquitin-conjugating and deubiquitinating enzymes (DUBs). Ubiquitin-specific protease 7 (USP7), a well-characterized DUB, plays multifaceted roles in various cellular processes, making it a promising therapeutic target. The plasticity of its catalytic domain and unique allosteric regulation by substrates or external or intramolecular factors facilitate the identification of highly selective USP7 inhibitors. These inhibitors can engage distinct ubiquitin-binding sites through covalent or non-covalent mechanisms. Despite its therapeutic promise, no USP7 inhibitors have entered clinical trials, underscoring the urgent need for novel therapeutics. Here we provide a crystallographic and functional landscape of USP7's multilayer regulation and analyze the structure-activity relationship of inhibitors by chemotypes. Additionally, we explore USP7's roles in diseases and discuss the challenges in USP7-targeted drug discovery and future directions for therapeutic development. This Perspective aims to provide a systematic overview of USP7, from its regulatory mechanisms to its therapeutic potential.
Collapse
Affiliation(s)
- Yihui Song
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fujian Medical University, Fuzhou, 350122, China
| | - Xiangli Ren
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Jinbo Xiong
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Wenwen Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Qianyan Zhao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Junbiao Chang
- College of Chemistry, Pingyuan Laboratory, Zhengzhou University, Zhengzhou 450001, China
| | - Bin Yu
- Tianjian Laboratory of Advanced Biomedical Sciences, Institute of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou 450001, China
- College of Chemistry, Pingyuan Laboratory, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
5
|
Parsaei S, Yaghoobi H, Beshkar P, Khonakdar Sangdehi HA, Khosravi Farsani MR, Safari O. Zingerone based green synthesized sodium doped zinc oxide nanoparticles eliminate U87 glioblastoma cells by inducing apoptosis. Sci Rep 2025; 15:13516. [PMID: 40251290 PMCID: PMC12008281 DOI: 10.1038/s41598-025-96962-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 04/01/2025] [Indexed: 04/20/2025] Open
Abstract
Grade IV astrocytoma, also referred to as glioblastoma (GBM), is the most common type of glioma, accounting for over 60% of all brain tumors. It is still a fatal illness in spite of years of investigation and does not currently have a treatment. Thus, scientists and medical professionals are constantly trying to understand the molecular processes and heterogeneity of GBM as well as looking for new ways to improve treatment results. Numerous studies have indicated that nanomaterials, and more especially nanoparticles, offer a great deal of potential for killing cancer cells; as a result, they are being considered as a potential alternative cancer treatment. Several studies have demonstrated that ZnO NPs have shown specific cytotoxicity against cancer cells while leaving normal cells unharmed. In this study we aim to synthesize sodium doped zinc oxide NPs using zingerone in an environmentally friendly manner to evaluate their cytotoxic effects on U87 GBM cell line and normal HEK cell line and investigate the occurrence of apoptosis via apoptosis assay by flowcytometry and gene expression study of TP53 and related genes to apoptosis and cell cycle regulation pathways. It was demonstrated that Na-doped ZnO NPs had a significant cytotoxic effect on U87 cells while having significantly less effect on normal HEK cells. Na-doped ZnO NPs eliminated cancerous cells through apoptosis induction and possibly cell cycle regulation via up-regulation of TP53, PTEN, BAX, P21 and down-regulation of Bcl2. The unique physicochemical properties of nanoparticles turn them into fascinating agents to treat GBM. Hence, the necessity of exploring the vast, yet unknown field of nanoparticles potentials cannot be over looked.
Collapse
Affiliation(s)
- Saman Parsaei
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Hajar Yaghoobi
- Clinical Biochemistry Research Center, Basic Health Science institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| | - Pezhman Beshkar
- Cellular and Molecular Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | | | | | - Omid Safari
- Cancer Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
6
|
Xie AX, Tansey W, Reznik E. UnitedMet harnesses RNA-metabolite covariation to impute metabolite levels in clinical samples. NATURE CANCER 2025:10.1038/s43018-025-00943-0. [PMID: 40251399 DOI: 10.1038/s43018-025-00943-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 03/06/2025] [Indexed: 04/20/2025]
Abstract
Comprehensively studying metabolism requires metabolite measurements. Such measurements, however, are often unavailable in large cohorts of tissue samples. To address this basic barrier, we propose a Bayesian framework ('UnitedMet') that leverages RNA-metabolite covariation to impute otherwise unmeasured metabolite levels from widely available transcriptomic data. UnitedMet is equally capable of imputing whole pool sizes and outcomes of isotope tracing experiments. We apply UnitedMet to investigate the metabolic impact of driver mutations in kidney cancer, identifying an association between BAP1 and a highly oxidative tumor phenotype. We similarly apply UnitedMet to determine that advanced kidney cancers upregulate oxidative phosphorylation relative to early-stage disease, that oxidative metabolism in kidney cancer is associated with inferior outcomes to anti-angiogenic therapy and that kidney cancer metastases demonstrate elevated oxidative phosphorylation. UnitedMet provides a scalable tool for assessing metabolic phenotypes when direct measurements are infeasible, facilitating unexplored avenues for metabolite-focused hypothesis generation.
Collapse
Affiliation(s)
- Amy X Xie
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Cornell University Weill Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Wesley Tansey
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Ed Reznik
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
7
|
Li L, Chai Q, Guo C, Wei J, Qin Y, Liu H, Lu Z. METTL3-mediated N6-methyladenosine modification contributes to vascular calcification. J Mol Cell Cardiol 2025; 203:22-34. [PMID: 40222552 DOI: 10.1016/j.yjmcc.2025.04.006] [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: 10/29/2024] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 04/15/2025]
Abstract
AIM Vascular calcification (VC) is a major adverse cardiovascular event in chronic kidney disease (CKD) patients. N6-methyladenosine (m6A) modification is vital for many biological processes, but its function and possible molecular mechanisms in VC are poorly understood. This study aimed to clarify the function and molecular mechanisms of N6-adenosine-methyltransferase-like 3 (METTL3) in VC. METHODS AND RESULTS The results of the bioinformatic analysis showed that METTL3 expression was significantly upregulated in calcified VSMCs. This finding was corroborated by phosphate-induced VSMCs calcification models and 5/6 nephrectomy-induced CKD mouse VC models. Afterward, Alizarin Red S staining and m6A dot blot analysis demonstrated METTL3 overexpression elevated m6A levels and encouraged calcification in VSMCs and mouse aortic rings, while METTL3 knockdown decreased m6A levels and inhibited calcium deposition in these experimental models. Furthermore, METTL3 promoted VC via the PTEN/AKT pathway, and MeRIP verified that METTL3 induced PTEN mRNA degradation by modifying it with m6A. In addition, molecular docking simulations and DARTS assays revealed that quercetin is a natural small-molecule inhibitor of METTL3. The current investigation demonstrated that quercetin mitigated VC by reducing METTL3-dependent m6A levels in vivo and in vitro. CONCLUSION In conclusion, this study unraveled the pathogenic mechanism of METTL3-mediated m6A modification in VC and provided new insights to establish METTL3 as a therapeutic target for VC.
Collapse
Affiliation(s)
- Long Li
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China; Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Quanyou Chai
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China; Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Chunling Guo
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Junyi Wei
- Department of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Yuqiao Qin
- Department of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Huimin Liu
- Department of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, China; Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China.
| | - Zhaoyang Lu
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou, China; Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou, China; Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
8
|
Mottaghi S, Abbaszadeh H, Valizadeh A, Hafezi K. The polyphenolic compound, α-conidendrin, exerts anti-colon cancer and anti-angiogenic effects by targeting several signaling molecules. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04090-2. [PMID: 40208320 DOI: 10.1007/s00210-025-04090-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 03/21/2025] [Indexed: 04/11/2025]
Abstract
Our previous study indicated that α-conidendrin had considerable anti-proliferative activities against breast cancer cell lines. The present study aimed to evaluate the anti-colon cancer and anti-angiogenic influences of α-conidendrin as well as its molecular mechanisms. The findings of the current study demonstrate that α-conidendrin possesses potent anti-colon cancer and anti-angiogenic effects. α-Conidendrin significantly inhibited the proliferation of colon cancer cells. This polyphenolic compound induced caspase-mediated apoptosis in HT-29 cells by modulating the PTEN/PI3K/Akt/mTOR signaling pathway. α-Conidendrin markedly upregulated the protein expression of PTEN and downregulated the protein expression of p-PI3K, p-AKt, and p-mTOR. The protein expression of caspase-3 and caspase-9 enhanced in colon cancer cells following treatment with α-conidendrin. This study also revealed the anti-angiogenic activities of α-conidendrin in the ex vivo and in vitro models. α-Conidendrin significantly downregulated the mRNA expression of HIF-1α, VEGF, MMP-2, and MMP-9 in endothelial cells. These data highlight that α-conidendrin can act as a novel and promising anti-cancer and anti-angiogenic agent for treatment of colon cancer.
Collapse
Affiliation(s)
- Sayeh Mottaghi
- Department of Pediatrics, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hassan Abbaszadeh
- Department of Pharmacology, Faculty of Pharmacy, Medicinal Plants Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Armita Valizadeh
- Department of Anatomical Sciences, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Katayoon Hafezi
- Department of Pharmacology, Faculty of Pharmacy, Medicinal Plants Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
9
|
Tu X, Lin W, Zhai X, Liang S, Huang G, Wang J, Jia W, Li S, Li B, Cheng B. Oleanolic acid inhibits M2 macrophage polarization and potentiates anti-PD-1 therapy in hepatocellular carcinoma by targeting miR-130b-3p-PTEN-PI3K-Akt signaling and glycolysis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156750. [PMID: 40250003 DOI: 10.1016/j.phymed.2025.156750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 03/26/2025] [Accepted: 04/08/2025] [Indexed: 04/20/2025]
Abstract
BACKGROUND Hypoxia promotes M2 polarization of macrophages and the formation of the immunosuppressive tumor microenvironment (TME) in hepatocellular carcinoma (HCC). Oleanolic acid (OA) has shown great potential in the treatment of HCC. However, the mechanisms of macrophage M2 polarization in hypoxic tumor TME and the regulating effect of OA is still unclear. OBJECTIVE To investigate the mechanisms of macrophage M2 polarization induced by hypoxic HCC cells-derived exosomes and examine the efficacy of OA in remedying the immunosuppressive TME and the anti-PD1 therapy potential. METHODS Hypoxic and normoxic HCC-derived exosomes (H-Exo and N-Exo) were collected by centrifugation. The microRNAs (miRNA) carried by the exosomes were sequenced and then screened to identify the functional miRNA. THP-1-induced macrophages were treated with exosomes or miRNAs to induce the M2 polarization of macrophages. Real-time RT-PCR and Western blotting were used to identify the direct target of miR-130b-3p and its downstream molecules. Hepa1-6 hepatoma-bearing mice were subjected to determine the efficacy of OA in regulating the TME and the anti-PD1 therapy potential. RESULTS H-Exo promotes macrophage M2 polarization, and thereby accelerates the migration and epithelial-mesenchymal transition (EMT) of HCC cells. Exosomal miRNA sequencing and subsequent functional validation showed that miR-130b-3p was the mediator of H-Exo-induced macrophage M2 polarization. PTEN was identified as the target of miR-130b-3p, and downregulation of PTEN by miR-130b-3p led to the activation of PI3K/Akt signaling and macrophage M2 polarization. In addition, miR-130b-3p also enhanced the glycolysis. OA suppressed H-Exo and miR-130b-3p-induced macrophage M2 polarization, also inhibited miR-130b-3p-induced glycolysis. In vivo, OA treatment enhanced the efficacy of anti-PD1 antibody by decreasing the number of M2 macrophages and increasing the number of CD8+ T cells. CONCLUSION Our findings uncover a new mechanism of hypoxic HCC cells-induced M2 polarization of macrophages through exosomal miR-130b-3p-PTEN-PI3K-Akt signaling. The combination therapy of OA with anti-PD1 antibody may lead to substantial improvements of the immunotherapy efficacy and expand the beneficiaries.
Collapse
Affiliation(s)
- Xiaoyu Tu
- Faculty of Traditional Chinese Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China; Department of Rehabilitation Medicine and Physiotherapy, the First Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai 200433, China
| | - Wanfu Lin
- Faculty of Traditional Chinese Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China; Oncology Department of Traditional Chinese Medicine, the First Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai 200433, China
| | - Xiaofeng Zhai
- Faculty of Traditional Chinese Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China; Oncology Department of Traditional Chinese Medicine, the First Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai 200433, China
| | - Shufang Liang
- Faculty of Traditional Chinese Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China; Oncology Department of Traditional Chinese Medicine, the First Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai 200433, China
| | - Guokai Huang
- Faculty of Traditional Chinese Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China; Oncology Department of Traditional Chinese Medicine, the First Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai 200433, China
| | - Jingfang Wang
- Faculty of Traditional Chinese Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China; Oncology Department of Traditional Chinese Medicine, the First Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai 200433, China
| | - Wentao Jia
- Faculty of Traditional Chinese Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China; Oncology Department of Traditional Chinese Medicine, the First Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai 200433, China
| | - Shu Li
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, China.
| | - Bai Li
- Department of Rehabilitation Medicine and Physiotherapy, the First Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai 200433, China.
| | - Binbin Cheng
- Faculty of Traditional Chinese Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China; Oncology Department of Traditional Chinese Medicine, the First Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai 200433, China.
| |
Collapse
|
10
|
Slarve I, Wang Y, Ding Y, Niu X, Tang Q, Jia C, Tu T, Hong H, Zhang G, Gu Y, Xu Z, Skinner S, He L, Hua B, Nguyen P, Zhou Y, Chen L, Ashouri K, Martynova A, Nakhoul C, Rastegarpour A, Alachkar H, Lenz HJ, El-Khoueiry A, Sher L, Chopra S, Yuan L, Stiles BL. Isoform specific regulation of osteopontin by AKT2 in hepatocytes and livers. Cell Signal 2025; 132:111799. [PMID: 40216173 DOI: 10.1016/j.cellsig.2025.111799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2025] [Accepted: 04/08/2025] [Indexed: 04/18/2025]
Abstract
Elevated levels of osteopontin (OPN), an inflammatory cytokine, are correlated with chronic inflammatory conditions and liver cancer. In this study, we explored the regulation of OPN in liver and hepatocytes by AKT1 vs. AKT2, the two AKT isoforms expressed in hepatocytes and livers. Using a mouse model lacking PTEN (phosphatase and tensin homologue deleted on chromosome 10), the negative regulator of phosphatidylinositol 3-kinase (PI3K)/AKT signaling, expression of secreted phosphoprotein 1 (Spp1), the gene encoding OPN, was found to be the topmost significantly upregulated gene in the liver. Using an add-back experiment in hepatocytes isolated from these mice, we show that PTEN regulates the expression of Spp1 mRNA as well as OPN protein levels. Exploring how PTEN regulates the expression of Spp1/OPN, we investigated the differential roles of AKT1 vs. AKT2 using hepatocytes isolated from mice lacking each AKT isoform in the liver. We showed here that levels of Spp1/OPN in hepatocytes are lost with deletion of Akt2 but not Akt1. Deletion of Akt2 significantly attenuated both basal expression of OPN and its response to IGF-1 stimulation. AKT1 loss, on the other hand, permitted more robust induction of OPN by IGF-1 stimulation. Furthermore, mice lacking AKT2 and PTEN exhibit significantly lower OPN expression in the liver. Together, this study showed that OPN levels are regulated by the PI3K/AKT signal in hepatocytes and that AKT2 but not AKT1 is responsible for its induction in response to stimulation of the PI3K signaling pathway.
Collapse
Affiliation(s)
- Ielyzaveta Slarve
- Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA
| | - Yushan Wang
- Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA
| | - Yining Ding
- Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA
| | - Xiaoteng Niu
- Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA
| | - Qi Tang
- Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA
| | - Chengyou Jia
- Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA; Department of Nuclear Medicine, Central Laboratory for Medical Research, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Taojian Tu
- Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA
| | - Handan Hong
- Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA
| | - Guo Zhang
- Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA
| | - Yiwei Gu
- Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA
| | - Zifei Xu
- Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA
| | - Samantha Skinner
- Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA
| | - Lina He
- Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA
| | - Brittney Hua
- Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA
| | - Phillip Nguyen
- Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA
| | - Yiren Zhou
- Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA
| | - Lulu Chen
- Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA
| | - Karam Ashouri
- Division of Medical Oncology, Keck School of Medicine, University of Southern California, Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA
| | - Anastasia Martynova
- Division of Medical Oncology, Keck School of Medicine, University of Southern California, Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA
| | - Christina Nakhoul
- Division of Medical Oncology, Keck School of Medicine, University of Southern California, Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA
| | - Ali Rastegarpour
- Division of Medical Oncology, Keck School of Medicine, University of Southern California, Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA
| | - Houda Alachkar
- Department of Clinical Pharmacy, Mann School of Pharmacy, University of Southern California, Los Angeles, CA 90089, USA
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Keck School of Medicine, University of Southern California, Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA
| | - Anthony El-Khoueiry
- Division of Medical Oncology, Keck School of Medicine, University of Southern California, Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA
| | - Linda Sher
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Shefali Chopra
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Liyun Yuan
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Bangyan Li Stiles
- Pharmacology and Pharmaceutical Sciences, Mann School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA; Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
11
|
Andrieu GP, Hypolite G, Latiri M, Balducci E, Costa C, Verhoeyen E, Courgeon M, Allatif O, Nemazanyy I, Panasyuk G, Wellen K, Herranz D, Genestier L, Macintyre E, Asnafi V, Tesio M. ATP citrate lyase is an essential player in the metabolic rewiring induced by PTEN loss during T-ALL development. Blood Adv 2025; 9:1670-1691. [PMID: 39546747 PMCID: PMC11999213 DOI: 10.1182/bloodadvances.2024013762] [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: 05/24/2024] [Revised: 10/17/2024] [Accepted: 10/30/2024] [Indexed: 11/17/2024] Open
Abstract
ABSTRACT Alterations inactivating the tumor suppressor gene PTEN drive the development of solid and hematologic cancers, such as T-cell acute lymphoblastic leukemia (T-ALL), in which phosphatase and tensin homolog (PTEN) loss defines poor-prognosis patients. We investigated the metabolic rewiring induced by PTEN loss in T-ALL, aiming to identify novel metabolic vulnerabilities. We showed that the enzyme adenosine triphosphate (ATP) citrate lyase (ACLY) is strictly required for the transformation of thymic immature progenitors and the growth of human T-ALL, which remain dependent on ACLY activity even upon transformation. Although Pten-mutant mice all died within 17 weeks, the concomitant Acly deletion prevented disease initiation in 70% of the animals. In these animals, ACLY promoted B-cell lymphoma (BCL-2) epigenetic upregulation and prevented the apoptosis of premalignant double-positive thymocytes. Transcriptomic and metabolic analysis of primary T-ALL cells next translated our findings to the human pathology, showing that PTEN-altered T-ALL cells activate ACLY and are sensitive to its genetic targeting. ACLY activation thus represents a metabolic vulnerability with therapeutic potential for high-risk patients with T-ALL.
Collapse
Affiliation(s)
- Guillaume P. Andrieu
- Laboratory of Onco-Hematology, Institut Necker Enfants Malades, and Institut national de la santé et de la recherche médicale U1115, Paris, France
- Université Paris-Cité, Paris, France
| | - Guillaume Hypolite
- Laboratory of Onco-Hematology, Institut Necker Enfants Malades, and Institut national de la santé et de la recherche médicale U1115, Paris, France
- Université Paris-Cité, Paris, France
| | - Mehdi Latiri
- Laboratory of Onco-Hematology, Institut Necker Enfants Malades, and Institut national de la santé et de la recherche médicale U1115, Paris, France
- Université Paris-Cité, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Necker Enfants Malades, Paris, France
| | - Estelle Balducci
- Laboratory of Onco-Hematology, Institut Necker Enfants Malades, and Institut national de la santé et de la recherche médicale U1115, Paris, France
- Université Paris-Cité, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Necker Enfants Malades, Paris, France
| | - Caroline Costa
- Vectorology Platform, International Center for Infectiology Research, Institut national de la santé et de la recherche médicale U1111, Lyon, France
- Université de Lyon 1, Lyon, France
- Centre national de la recherche sciéntifique UMR5308, Lyon, France
- Ecole Normale Supérieure de Lyon, Lyon, France
| | - Els Verhoeyen
- Laboratory of Metabolic control of cellular death, Centre Méditerranéen de Médecine Moléculaire, Institut national de la santé et de la recherche médicale U1065, Nice, France
| | - Marianne Courgeon
- Laboratory of Onco-Hematology, Institut Necker Enfants Malades, and Institut national de la santé et de la recherche médicale U1115, Paris, France
- Université Paris-Cité, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Necker Enfants Malades, Paris, France
| | - Omran Allatif
- Platform Bioinformatics-Biostatistics, Centre International de Recherche en Infectiologie, Institut national de la santé et de la recherche médicale U1111, Lyon, France
| | - Ivan Nemazanyy
- Platform for Metabolic Analyses, Structure Fédérative de Recherche Necker, Institut national de la santé et de la recherche médicale US24, Paris, France
- Centre national de la recherche sciéntifique, unité de recherche associé 3633, Paris, France
| | - Ganna Panasyuk
- Laboratory of Nutrient Sensing Mechanisms, Institut Necker Enfants Malades, Institut national de la santé et de la recherche médicale U1151, Paris, France
- Centre national de la recherche sciéntifique, unité mixte de recherche 8253, Paris, France
| | - Kathryn Wellen
- Abramson Family Cancer Research Institute, University of Pennsylvania, Department of Cancer Biology, Perelman School of Medicine, Philadelphia, PA
| | - Daniel Herranz
- Rutgers Cancer Institute, Department of Pharmacology, Department of Pediatrics, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ
| | - Laurent Genestier
- Université de Lyon 1, Lyon, France
- Lymphoma Immune-biology, Centre International de Recherche en Infectiologie, Institut national de la santé et de la recherche médicale U1111, Lyon, France
- Centre national de la recherche sciéntifique, unité mixte de recherche 5308, Lyon, France
| | - Elizabeth Macintyre
- Laboratory of Onco-Hematology, Institut Necker Enfants Malades, and Institut national de la santé et de la recherche médicale U1115, Paris, France
- Université Paris-Cité, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Necker Enfants Malades, Paris, France
| | - Vahid Asnafi
- Laboratory of Onco-Hematology, Institut Necker Enfants Malades, and Institut national de la santé et de la recherche médicale U1115, Paris, France
- Université Paris-Cité, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Necker Enfants Malades, Paris, France
| | - Melania Tesio
- Laboratory of Onco-Hematology, Institut Necker Enfants Malades, and Institut national de la santé et de la recherche médicale U1115, Paris, France
- Université de Lyon 1, Lyon, France
- Lymphoma Immune-biology, Centre International de Recherche en Infectiologie, Institut national de la santé et de la recherche médicale U1111, Lyon, France
- Centre national de la recherche sciéntifique, unité mixte de recherche 5308, Lyon, France
| |
Collapse
|
12
|
Thabet EH, Khalil NA, Essawy MM, Harby SA, Solaiman AA, El Gazaerly HM, Khalifa YH. MiRNA-21-loaded chitosan nanoparticles ameliorate pancreatic apoptosis and oxidative stress in diabetic rats. Life Sci 2025; 366-367:123471. [PMID: 39956187 DOI: 10.1016/j.lfs.2025.123471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 02/09/2025] [Accepted: 02/13/2025] [Indexed: 02/18/2025]
Abstract
BACKGROUND Accelerated Pancreatic β-cell apoptosis and oxidative stress are the mainstays of type-1 diabetes. MicroRNA-21's (miRNA-21) role in regulating pancreatic β-cell function remains indefinable. MATERIAL AND METHODS Five groups of rats were used in this study (healthy controls (Ia), controls that received only chitosan (CS) nanoparticles (NPs)(Ib), streptozotocin (STZ)-induced diabetics rats (II),STZ-induced diabetic rats that received only CS-NPs(III), and STZ-induced diabetic rats treated with mi-RNA-21-CS-NPs(IV). Sera were collected for measurement of fasting blood glucose levels (FBG), insulin, oxidative stress, and intraperitoneal glucose intolerance tests. Pancreatic tissue was collected after sacrifice partly for histological examination and for oxidative stress assessment and evaluation of PTEN/ AKT using qRT-PCR. KEY FINDINGS We showed over-expression of cleaved-caspase-3 indicating accelerated apoptosis in the β-cell of STZ-induced diabetic rats. Apoptosis was significantly ameliorated by miRNA-21-CS. MiRNA-21-CS-NPs faithfully restored serum fasting insulin, and FBG, and reduced serum and pancreatic oxidative stress markers while enhancing the total antioxidant capacity. Histological examination revealed that miRNA-21 restored healthy β-cell architecture, decreased cleaved-caspase-3, and increased insulin secretion. Transmission electron microscopy revealed increased mitochondrial circularity that significantly correlated with an exaggerated oxidative stress profile as shown by high serum and pancreatic malondialdehyde (MDA), low glutathione peroxidase, and total antioxidant capacity in STZ-induced diabetes. This oxidative profile was reversed using miRNA-21-CS-NPs. Mi-RNA-21 therapy downregulated PTEN but increased AKT and pAKT expression. Altogether, we show that miRNA-21 restored normal islet β-cell structure and insulin secretion through PTEN inhibition. SIGNIFICANCE miRNA-21- CS-NPs are promising targeted therapeutics that may effectively decrease the global burden of diabetes.
Collapse
Affiliation(s)
- Eman H Thabet
- Department of Medical Physiology, Faculty of Medicine, University of Alexandria, 21500 Alexandria, Egypt; Center of Excellence for Research in Regenerative Medicine and Applications (CERRMA), Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt.
| | - Nehal A Khalil
- Department of Medical Biochemistry, Faculty of Medicine, University of Alexandria, 21500 Alexandria, Egypt; Center of Excellence for Research in Regenerative Medicine and Applications (CERRMA), Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Marwa M Essawy
- Oral Pathology Department, Faculty of Dentistry, Alexandria University, 21500 Alexandria, Egypt; Center of Excellence for Research in Regenerative Medicine and Applications (CERRMA), Faculty of Medicine, Alexandria University, Alexandria 21500, Egypt
| | - Sahar A Harby
- Department of Clinical Pharmacology, Faculty of Medicine, University of Alexandria, 21500 Alexandria, Egypt
| | - Amany A Solaiman
- Department of Histology and Cell Biology, Faculty of Medicine, University of Alexandria, 21500 Alexandria, Egypt
| | - Hanaa M El Gazaerly
- Department of Oral Pathology, Faculty of Dentistry, Tanta University, Tanta 31527, Egypt
| | - Yassmin H Khalifa
- Department of Histology and Cell Biology, Faculty of Medicine, University of Alexandria, 21500 Alexandria, Egypt
| |
Collapse
|
13
|
Hashemi M, Fard AA, Pakshad B, Asheghabadi PS, Hosseinkhani A, Hosseini AS, Moradi P, Mohammadbeygi Niye M, Najafi G, Farahzadi M, Khoushab S, Taheriazam A, Farahani N, Mohammadi M, Daneshi S, Nabavi N, Entezari M. Non-coding RNAs and regulation of the PI3K signaling pathway in lung cancer: Recent insights and potential clinical applications. Noncoding RNA Res 2025; 11:1-21. [PMID: 39720352 PMCID: PMC11665378 DOI: 10.1016/j.ncrna.2024.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 11/11/2024] [Accepted: 11/21/2024] [Indexed: 12/26/2024] Open
Abstract
Lung cancer (LC) is one of the most common causes of cancer-related death worldwide. It has been demonstrated that the prognosis of current drug treatments is affected by a variety of factors, including late stage, tumor recurrence, inaccessibility to appropriate treatments, and, most importantly, chemotherapy resistance. Non-coding RNAs (ncRNAs) contribute to tumor development, with some acting as tumor suppressors and others as oncogenes. The phosphoinositide 3-kinase (PI3Ks)/AKT serine/threonine kinase pathway is one of the most important common targets of ncRNAs in cancer, which is widely applied to modulate the cell cycle and a variety of biological processes, including cell growth, mobility survival, metabolic activity, and protein production. Discovering the biology of ncRNA-PI3K/AKT signaling may lead to advances in cancer diagnosis and treatment. As a result, we investigated the expression and role of PI3K/AKT-related ncRNAs in clinical characteristics of lung cancer, as well as their functions as potential biomarkers in lung cancer diagnosis, prognosis, and treatment.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Asal Abolghasemi Fard
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Bita Pakshad
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Pezhman Shafiei Asheghabadi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Amineh Hosseinkhani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Atena Sadat Hosseini
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Parham Moradi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mohammadreza Mohammadbeygi Niye
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Ghazal Najafi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mohadeseh Farahzadi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saloomeh Khoushab
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mahya Mohammadi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Salman Daneshi
- Department of Public Health, School of Health, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia, V8V 1P7, Canada
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| |
Collapse
|
14
|
Banjan B, Vishwakarma R, Ramakrishnan K, Dev RR, Kalath H, Kumar P, Soman S, Raju R, Revikumar A, Rehman N, Abhinand CS. Targeting AFP-RARβ complex formation: a potential strategy for treating AFP-positive hepatocellular carcinoma. Mol Divers 2025; 29:1337-1352. [PMID: 38955977 DOI: 10.1007/s11030-024-10915-8] [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: 03/21/2024] [Accepted: 06/09/2024] [Indexed: 07/04/2024]
Abstract
Alpha-fetoprotein (AFP) is a glycoprotein primarily expressed during embryogenesis, with declining levels postnatally. Elevated AFP levels correlate with pathological conditions such as liver fibrosis, cirrhosis, and hepatocellular carcinoma (HCC). Recent investigations underscore AFP's intracellular role in HCC progression, wherein it forms complexes with proteins like Phosphatase and tensin homolog (PTEN), Caspase 3 (CASP3), and Retinoic acid receptors and Retinoid X receptors (RAR/RXR). RAR and RXR regulate gene expression linked to cell death and tumorigenesis in normal physiology. AFP impedes RAR/RXR dimerization, nuclear translocation, and function, promoting gene expression favoring cancer progression in HCC that provoked us to target AFP as a drug candidate. Despite extensive studies, inhibitors targeting AFP to disrupt complex formation and activities remain scarce. In this study, employing protein-protein docking, amino acid residues involved in AFP-RARβ interaction were identified, guiding the definition of AFP's active site for potential inhibitor screening. Currently, kinase inhibitors play a significant role in cancer treatment and, the present study explores the potential of repurposing FDA-approved protein kinase inhibitors to target AFP. Molecular docking with kinase inhibitors revealed Lapatinib as a candidate drug of the AFP-RARβ complex. Molecular dynamics simulations and binding energy calculations, employing Mechanic/Poisson-Boltzmann Surface Area (MM-PBSA), confirmed Lapatinib's stability with AFP. The study suggests Lapatinib's potential in disrupting the AFP-RARβ complex, providing a promising avenue for treating molecularly stratified AFP-positive HCC or its early stages.
Collapse
Affiliation(s)
- Bhavya Banjan
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to Be University), Mangalore, 575018, India
| | - Riya Vishwakarma
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to Be University), Mangalore, 575018, India
| | - Krishnapriya Ramakrishnan
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to Be University), Mangalore, 575018, India
| | - Radul R Dev
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to Be University), Mangalore, 575018, India
| | - Haritha Kalath
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to Be University), Mangalore, 575018, India
| | - Pankaj Kumar
- Nitte (Deemed to Be University), Department of Pharmaceutical Chemistry, NGSMPS, NGSM Institute of Pharmaceutical Sciences, Mangalore, 575018, Karnataka, India
| | - Sowmya Soman
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to Be University), Mangalore, 575018, India
| | - Rajesh Raju
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to Be University), Mangalore, 575018, India
- Centre for Systems Biology and Molecular Medicine (CSBMM), Yenepoya Research Centre, Yenepoya (Deemed to Be University), Deralakatte, Mangalore, 575018, Karnataka, India
| | - Amjesh Revikumar
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to Be University), Mangalore, 575018, India
- Kerala Genome Data Centre, Kerala Development and Innovation Strategic Council, Vazhuthacaud, Thiruvananthapuram, Kerala, 695014, India
| | - Niyas Rehman
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to Be University), Mangalore, 575018, India.
| | - Chandran S Abhinand
- Centre for Systems Biology and Molecular Medicine (CSBMM), Yenepoya Research Centre, Yenepoya (Deemed to Be University), Deralakatte, Mangalore, 575018, Karnataka, India.
| |
Collapse
|
15
|
Li Q, Cheng J, Qin D, Xiao S, Yao C. Exosomal miR-92b-5p regulates N4BP1 to enhance PTEN mono-ubiquitination in doxorubicin-resistant AML. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2025; 8:16. [PMID: 40201312 PMCID: PMC11977356 DOI: 10.20517/cdr.2024.140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 03/04/2025] [Accepted: 03/25/2025] [Indexed: 04/10/2025]
Abstract
Aim: Doxorubicin, pivotal for acute myeloid leukemia (AML) treatment, often succumbs to resistance, impeding therapeutic success. Although exosomal transfer is linked to chemoresistance, the detailed role of exosomal miRNAs in doxorubicin resistance remains incompletely understood. Methods: We employed miRNA sequencing to delineate the profile of exosomal miRNAs in doxorubicin-resistant K562/DOX cells and AML patients. Subsequently, qPCR was utilized to scrutinize the expression of exosomal miR-92b-5p in these resistant cells and AML patients. A dual-luciferase reporter assay was conducted to elucidate the direct binding of miR-92b-5p to NEDD4 binding protein 1 (N4BP1). Furthermore, interactions between N4BP1 and NEDD4, as well as between NEDD4 and PTEN, were investigated by co-immunoprecipitation (Co-IP). Meanwhile, the ubiquitination of PTEN was also examined by Co-IP. Western blot analysis was applied to assess the expression levels of N4BP1, NEDD4, PTEN, RAD51, and proteins associated with the PI3K-AKT-mTOR pathway. Gain- and loss-of-function studies were conducted to ascertain the functional role of miR-92b-5p in doxorubicin resistance by using miR-92b-5p-mimic and miR-92b-5p-inhibitor transfections. Results: Our study found exosomal miR-92b-5p was upregulated both in doxorubicin-resistant cells and AML patients. Moreover, miR-92b-5p targets N4BP1, promoting NEDD4-mediated mono-ubiquitination of PTEN. This alters PTEN's subcellular localization, promoting nuclear PTEN and reducing cytoplasmic PTEN, which in turn leads to increased RAD51 for DNA repair and activation of the PI3K-AKT-mTOR pathway for cell proliferation, contributing to doxorubicin resistance. Conclusion: Our study reveals a novel mechanism of doxorubicin resistance mediated by exosomal miR-92b-5p and provides potential therapeutic targets for overcoming drug resistance in AML.
Collapse
Affiliation(s)
- Qianyuan Li
- Department of General Medicine, The 3rd Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
- Department of Hematology, The 3rd Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Jie Cheng
- Department of Hematology, The 3rd Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Danni Qin
- Department of Hematology, The 3rd Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Sheng Xiao
- Department of Pathology, The 3rd Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Chenjiao Yao
- Department of General Medicine, The 3rd Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
- Department of Hematology, The First Affiliated Hospital of Hainan Medical University, Haikou 570105, Hainan, China
| |
Collapse
|
16
|
Zhang Y, Chen X, Li J, Chen X, Zhao J, Liu Q, Li X, Wang X, Xiao Z. Seminal plasma exosome derived miR-26-5p can regulate decidual macrophage polarization via PTEN / PI3K / AKT signaling pathway. Sci Rep 2025; 15:9192. [PMID: 40097471 PMCID: PMC11914418 DOI: 10.1038/s41598-025-92880-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 03/03/2025] [Indexed: 03/19/2025] Open
Abstract
The immunomodulatory effects of seminal plasma (SP) on the maternal immune system play an important role in the implantation and development of the embryo. Decidual macrophages (dMΦs) are one of the major immune cells in the maternal-fetal immune microenvironment, and their M2-type polarization facilitates the establishment and maintenance of pregnancy. However, the role of SP on the polarization of dMΦs is unknown. In this study, we investigated the role and mechanism of SP on the polarization of dMΦs by gene chip sequencing as well as in vitro and in vivo experiments. The results revealed that SP promoted dMΦs M2-type polarization. Gene chip sequencing revealed that miR-26-5p was highly expressed in seminal exosomes (SEs) which could act on PTEN/PI3K/AKT signaling pathway and significantly promote MΦs M2 polarization. Moreover, SEs supplementation significantly reduced embryo resorption in spontaneously aborted mice. In conclusion, our study demonstrated that the SEs derived miR-26-5p in SP promoted the M2 polarization of dMΦs by targeting PTEN/PI3K/AKT signaling pathway, which created an immune-tolerant environment conducive to embryo implantation and development. This study provided new ideas for clinical SP-assisted therapy to improve pregnancy outcomes.
Collapse
Affiliation(s)
- Yan Zhang
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Xiaolin Chen
- Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Jie Li
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Xin Chen
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Jing Zhao
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Qing Liu
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Xiaoling Li
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Xinyu Wang
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Zhuoni Xiao
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| |
Collapse
|
17
|
Nisha, Thapliyal D, Gohil B, Modak AS, Singh NT, Mukherjee C, Ahuja S, Sahu BS, Singh MD. Downregulation of Pten Improves Huntington's Disease Phenotype by Reducing Htt Aggregates and Cell Death. Mol Neurobiol 2025:10.1007/s12035-025-04816-6. [PMID: 40042729 DOI: 10.1007/s12035-025-04816-6] [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: 03/27/2024] [Accepted: 02/26/2025] [Indexed: 03/18/2025]
Abstract
Huntington's disease (HD) is a dominantly inherited neurodegenerative disorder that stems from the expansion of CAG repeats within the coding region of Huntingtin (HTT) gene. Currently, there exists no effective therapeutic intervention that can prevent the progression of the disease. Our study aims to identify a novel genetic modifier with therapeutic potential. We employ transgenic flies containing HTT.ex1.Q93 and mRFP-HTT.588.Q138 constructs, which encode mutant pathogenic Huntingtin (Htt) proteins featuring 93 and 138 polyglutamine (Q) repeats respectively. The resultant mutant proteins cause the loss of photoreceptor neurons in the eye and a progressive loss of neuronal tissues in the brain and motor neurons in Drosophila. Several findings have demonstrated the association of HD with growth factor signaling defects. Phosphatase and tensin homolog (Pten) have been implicated in the negative regulation of the Insulin signaling/receptor tyrosine signaling pathway which regulates the growth and survival of cells. In the present study, we downregulated Pten and found a significant improvement in morphological phenotypes in the eye, brain, and motor neurons. These findings were further correlated with the enhancement of the functional vision and climbing ability of the flies. We also found the reduction in both Htt aggregate and caspase levels which are involved in the apoptotic pathway. In alignment with the genetic modulation of Pten, we elucidated the protective role of Pten inhibition through the utilization of VO-OHpic. VO-OHpic improved the climbing ability of flies and reduced the poly(Q) aggregates and apoptosis levels. A similar reduction in Htt aggregates was observed in the mouse neuronal inducible HD cell line model. Our study illustrates that Pten inhibition is a potential therapeutic approach for HD.
Collapse
Affiliation(s)
- Nisha
- National Brain Research Centre, NH-8, Manesar, Gurgaon, Haryana, 122052, India
| | - Deepti Thapliyal
- National Brain Research Centre, NH-8, Manesar, Gurgaon, Haryana, 122052, India
| | - Bhavya Gohil
- National Brain Research Centre, NH-8, Manesar, Gurgaon, Haryana, 122052, India
| | - Aninda Sundar Modak
- National Brain Research Centre, NH-8, Manesar, Gurgaon, Haryana, 122052, India
| | - N Tarundas Singh
- National Brain Research Centre, NH-8, Manesar, Gurgaon, Haryana, 122052, India
| | | | - Sanchi Ahuja
- National Brain Research Centre, NH-8, Manesar, Gurgaon, Haryana, 122052, India
| | | | | |
Collapse
|
18
|
Zhou J, Yang F, Zheng C, Chen Y, Chen M, Lin Q, Chang C, Cai S, Sun Z, Li H, Qin L, Chen Y. OncomiR mdv1-miR-M7-5p promotes avian lymphomatosis by modulating the BCL2/Bax mitochondrial apoptosis signaling pathway. Vet Microbiol 2025; 302:110417. [PMID: 39889336 DOI: 10.1016/j.vetmic.2025.110417] [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: 09/09/2024] [Revised: 01/22/2025] [Accepted: 01/27/2025] [Indexed: 02/03/2025]
Abstract
Marek's disease virus (MDV) is an oncogenic alphaherpesvirus that infects poultry and causes fatal lymphomas in infected chickens. Notably, the mdv1-miR-M7-5p, a pivotal oncomiR encoded by MDV, is closely associated with viral replication and latency. Here, mdv1-miR-M7-5p was transfected into the chicken lymphoma cell line MSB1, which resulted in the inhibition of lymphoma cell apoptosis and an increase in lymphoma cell proliferation and migration. Additionally, the expression of the tumor suppressor genes p53 and ARRDC3 were significantly downregulated, while the MDV latency-associated genes such as ICP4 and ICP27 were significantly upregulated. The BCL2/Bax ratio was increased while the expression of genes involved in the apoptotic signaling pathway were decreased. Furthermore, our mitochondrial function experiments in MSB1 cells demonstrated that mdv1-miR-M7-5p enhanced mitochondrial ATP release and altered the mitochondrial membrane potential, thereby affecting mitochondrial function and inhibiting lymphoma cell apoptosis. Dual-luciferase assays revealed that mdv1-miR-M7-5p binds to caspase-6. For the in vivo study, a cholesterol-modified inhibitor of mdv1-miR-M7-5p was administered to chickens. Inhibition of mdv1-miR-M7-5p resulted in a lower mortality rate than that in the control groups. Furthermore, the expression levels of the cytokines interferon-gamma (IFN-γ), interleukin (IL)-4, and IL-17 in the plasma of MDV-infected chickens were significantly increased. A marked increase was observed in apoptosis in the spleen tissues, and the expression of apoptosis-related genes including caspase-3 and tumor suppressor gene PTEN in immune organs, including the spleen, bursa of Fabricius, and thymus, were markedly upregulated. In summary, the oncogenic miRNA mdv1-miR-M7-5p promotes MDV latency and may facilitate lymphoma formation by mediating the BCL2/CytC signaling pathway. This mediation enhances mitochondrial function and inhibits lymphoma cell apoptosis, thereby contributing to lymphoma development.
Collapse
Affiliation(s)
- Jun Zhou
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Animal Science and Technology, Foshan University, Foshan 528225, China
| | - Fan Yang
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Animal Science and Technology, Foshan University, Foshan 528225, China
| | - Congsen Zheng
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Animal Science and Technology, Foshan University, Foshan 528225, China
| | - Yanting Chen
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Animal Science and Technology, Foshan University, Foshan 528225, China
| | - Meiting Chen
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Animal Science and Technology, Foshan University, Foshan 528225, China
| | - Qiaoer Lin
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Animal Science and Technology, Foshan University, Foshan 528225, China
| | - Chuanzhe Chang
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Animal Science and Technology, Foshan University, Foshan 528225, China
| | - Shikai Cai
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Animal Science and Technology, Foshan University, Foshan 528225, China
| | - Zhaoyang Sun
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Animal Science and Technology, Foshan University, Foshan 528225, China
| | - Hua Li
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Animal Science and Technology, Foshan University, Foshan 528225, China
| | - Limei Qin
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Animal Science and Technology, Foshan University, Foshan 528225, China.
| | - Yanfeng Chen
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Animal Science and Technology, Foshan University, Foshan 528225, China.
| |
Collapse
|
19
|
Nelson AB, Reese LE, Rono E, Queathem ED, Qiu Y, McCluskey BM, Crampton A, Conniff E, Cummins K, Boytim E, Dansou S, Hwang J, Safo SE, Puchalska P, Wood DK, Schwertfeger KL, Crawford PA. Deciphering Colorectal Cancer-Hepatocyte Interactions: A Multiomics Platform for Interrogation of Metabolic Crosstalk in the Liver-Tumor Microenvironment. Int J Mol Sci 2025; 26:1976. [PMID: 40076609 PMCID: PMC11900982 DOI: 10.3390/ijms26051976] [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: 12/30/2024] [Revised: 02/01/2025] [Accepted: 02/18/2025] [Indexed: 03/14/2025] Open
Abstract
Metabolic reprogramming is a hallmark of cancer, enabling tumor cells to adapt to and exploit their microenvironment for sustained growth. The liver is a common site of metastasis, but the interactions between tumor cells and hepatocytes remain poorly understood. In the context of liver metastasis, these interactions play a crucial role in promoting tumor survival and progression. This study leverages multiomics coverage of the microenvironment via liquid chromatography and high-resolution, high-mass-accuracy mass spectrometry-based untargeted metabolomics, 13C-stable isotope tracing, and RNA sequencing to uncover the metabolic impact of co-localized primary hepatocytes and a colon adenocarcinoma cell line, SW480, using a 2D co-culture model. Metabolic profiling revealed disrupted Warburg metabolism with an 80% decrease in glucose consumption and 94% decrease in lactate production by hepatocyte-SW480 co-cultures relative to SW480 control cultures. Decreased glucose consumption was coupled with alterations in glutamine and ketone body metabolism, suggesting a possible fuel switch upon co-culturing. Further, integrated multiomics analysis indicates that disruptions in metabolic pathways, including nucleoside biosynthesis, amino acids, and TCA cycle, correlate with altered SW480 transcriptional profiles and highlight the importance of redox homeostasis in tumor adaptation. Finally, these findings were replicated in three-dimensional microtissue organoids. Taken together, these studies support a bioinformatic approach to study metabolic crosstalk and discovery of potential therapeutic targets in preclinical models of the tumor microenvironment.
Collapse
Affiliation(s)
- Alisa B. Nelson
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA (E.D.Q.)
| | - Lyndsay E. Reese
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA; (L.E.R.)
| | - Elizabeth Rono
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA; (E.R.); (S.D.)
| | - Eric D. Queathem
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA (E.D.Q.)
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Yinjie Qiu
- Minnesota Supercomputing Institute, Minneapolis, MN 55455, USA
| | | | - Alexandra Crampton
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA; (E.R.); (S.D.)
| | - Eric Conniff
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA; (E.R.); (S.D.)
| | - Katherine Cummins
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA; (E.R.); (S.D.)
| | - Ella Boytim
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Senali Dansou
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA; (E.R.); (S.D.)
| | - Justin Hwang
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Sandra E. Safo
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, MN 55455, USA
| | - Patrycja Puchalska
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA (E.D.Q.)
| | - David K. Wood
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA; (E.R.); (S.D.)
| | - Kathryn L. Schwertfeger
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA; (L.E.R.)
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Peter A. Crawford
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA (E.D.Q.)
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
20
|
Saunders RA, Allen WE, Pan X, Sandhu J, Lu J, Lau TK, Smolyar K, Sullivan ZA, Dulac C, Weissman JS, Zhuang X. A platform for multimodal in vivo pooled genetic screens reveals regulators of liver function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.18.624217. [PMID: 39605605 PMCID: PMC11601512 DOI: 10.1101/2024.11.18.624217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Organ function requires coordinated activities of thousands of genes in distinct, spatially organized cell types. Understanding the basis of emergent tissue function requires approaches to dissect the genetic control of diverse cellular and tissue phenotypes in vivo. Here, we develop paired imaging and sequencing methods to construct large-scale, multi-modal genotype-phenotypes maps in tissue with pooled genetic perturbations. Using imaging, we identify genetic perturbations in individual cells while simultaneously measuring their gene expression and subcellular morphology. Using single-cell sequencing, we measure transcriptomic responses to the same genetic perturbations. We apply this approach to study hundreds of genetic perturbations in the mouse liver. Our study reveals regulators of hepatocyte zonation and liver unfolded protein response, as well as distinct pathways that cause hepatocyte steatosis. Our approach enables new ways of interrogating the genetic basis of complex cellular and organismal physiology and provides crucial training data for emerging machine-learning models of cellular function.
Collapse
Affiliation(s)
- Reuben A. Saunders
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Whitehead Institute, Cambridge, MA 02139, USA
- University of California, San Francisco, San Francisco, CA 94158, USA
- Present address: Society of Fellows, Harvard University, MA 02138, USA
- These authors contributed equally
| | - William E. Allen
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Society of Fellows, Harvard University, Cambridge, MA 02138, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Present address: Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305; Arc Institute, Palo Alto, CA 94304
- These authors contributed equally
- Lead contact
| | - Xingjie Pan
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Lead AI Scientist
| | - Jaspreet Sandhu
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Whitehead Institute, Cambridge, MA 02139, USA
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jiaqi Lu
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Thomas K. Lau
- Department of Statistics, Stanford University, Stanford, CA 94305
| | - Karina Smolyar
- Whitehead Institute, Cambridge, MA 02139, USA
- Department of Biology, MIT, Cambridge, MA 02139 USA
| | - Zuri A. Sullivan
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Catherine Dulac
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Jonathan S. Weissman
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Whitehead Institute, Cambridge, MA 02139, USA
- Department of Biology, MIT, Cambridge, MA 02139 USA
| | - Xiaowei Zhuang
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Department of Physics, Harvard University, Cambridge, MA 02138, USA
| |
Collapse
|
21
|
Bao Y, Ma Y, Huang W, Bai Y, Gao S, Xiu L, Xie Y, Wan X, Shan S, Chen C, Qu L. Regulation of autophagy and cellular signaling through non-histone protein methylation. Int J Biol Macromol 2025; 291:139057. [PMID: 39710032 DOI: 10.1016/j.ijbiomac.2024.139057] [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: 09/17/2024] [Revised: 12/06/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
Autophagy is a highly conserved catabolic pathway that is precisely regulated and plays a significant role in maintaining cellular metabolic balance and intracellular homeostasis. Abnormal autophagy is directly linked to the development of various diseases, particularly immune disorders, neurodegenerative conditions, and tumors. The precise regulation of proteins is crucial for proper cellular function, and post-translational modifications (PTMs) are key epigenetic mechanisms in the regulation of numerous biological processes. Multiple proteins undergo PTMs that influence autophagy regulation. Methylation modifications on non-histone lysine and arginine residues have been identified as common PTMs critical to various life processes. This paper focused on the regulatory effects of non-histone methylation modifications on autophagy, summarizing related research on signaling pathways involved in autophagy-related non-histone methylation, and discussing current challenges and clinical significance. Our review concludes that non-histone methylation plays a pivotal role in the regulation of autophagy and its associated signaling pathways. Targeting non-histone methylation offers a promising strategy for therapeutic interventions in diseases related to autophagy dysfunction, such as cancer and neurodegenerative disorders. These findings provide a theoretical basis for the development of non-histone-methylation-targeted drugs for clinical use.
Collapse
Affiliation(s)
- Yongfen Bao
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, Xianning 437000, China; School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning 437000, China
| | - Yaoyao Ma
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, Xianning 437000, China; School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning 437000, China
| | - Wentao Huang
- Department of Physiology, Hunan Normal University School of Medicine, Changsha 410013, China
| | - Yujie Bai
- Department of Scientific Research and Education, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330000, China
| | - Siying Gao
- Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Luyao Xiu
- Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Yuyang Xie
- Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Xinrong Wan
- Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Shigang Shan
- School of Public Health and Nursing, Hubei University of Science and Technology, Hubei 437000, China
| | - Chao Chen
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Lihua Qu
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, Xianning 437000, China; School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning 437000, China.
| |
Collapse
|
22
|
Peter RM, Sarwar MS, Wang L, Chou P, Wang C, Wang Y, Su X, Kong AN. Dietary phytochemical indole-3-carbinol regulates metabolic reprogramming in mouse prostate tissue. Pharm Res 2025; 42:237-247. [PMID: 39904853 PMCID: PMC11880055 DOI: 10.1007/s11095-025-03820-8] [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: 10/09/2024] [Accepted: 01/08/2025] [Indexed: 02/06/2025]
Abstract
PURPOSE Indole-3-carbinol (I3C) is shown to possess multiple pharmacological activities such as anti-inflammatory, antimicrobial, antioxidant, antiviral, and anti-cancer activities. It is widely accepted as modulator of multiple signaling pathways particularly those related to cell cycle, cell growth and division, angiogenesis, apoptosis and immunity. We explored the metabolic reprogramming based on treatment with I3C in mice prostate tissue. METHODS In this study we utilized Pten knockout (KO)-induced prostate tumorigenesis mouse model to examine mechanism of action of I3C via metabolic rewiring. Phosphatase and tensin homolog deleted on chromosome 10 (Pten), a tumor suppressor gene is frequently found to be mutated or deleted in prostate cancer. Untargeted metabolomics was performed using liquid-chromatography mass-spectrometry (LC-MS) based platform to investigate Pten-dependent and Pten-independent metabolic targets of I3C. RESULTS The most impacted pathways by I3C included pyrimidine metabolism, arginine and proline metabolism, porphyrin metabolism, citrate cycle and lipoic acid metabolism. CONCLUSION These pathways taken together help in understanding the overall health beneficial effects of I3C.
Collapse
Affiliation(s)
- Rebecca Mary Peter
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, The State University of New Jersey, RutgersPiscataway, NJ, 08854, USA
- Graduate Program of Pharmaceutical Sciences, Ernest Mario School of Pharmacy, The State University of New Jersey, RutgersPiscataway, NJ, 08854, USA
| | - Md Shahid Sarwar
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, The State University of New Jersey, RutgersPiscataway, NJ, 08854, USA
| | - Lujing Wang
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, The State University of New Jersey, RutgersPiscataway, NJ, 08854, USA
- Graduate Program of Pharmaceutical Sciences, Ernest Mario School of Pharmacy, The State University of New Jersey, RutgersPiscataway, NJ, 08854, USA
| | - Pochung Chou
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, The State University of New Jersey, RutgersPiscataway, NJ, 08854, USA
- Graduate Program of Pharmaceutical Sciences, Ernest Mario School of Pharmacy, The State University of New Jersey, RutgersPiscataway, NJ, 08854, USA
| | - Chao Wang
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, The State University of New Jersey, RutgersPiscataway, NJ, 08854, USA
| | - Yujue Wang
- Metabolomics Shared Resource, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08901, USA
- Department of Medicine, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA
| | - Xiaoyang Su
- Metabolomics Shared Resource, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08901, USA
- Department of Medicine, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA
| | - Ah-Ng Kong
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, The State University of New Jersey, RutgersPiscataway, NJ, 08854, USA.
- Graduate Program of Pharmaceutical Sciences, Ernest Mario School of Pharmacy, The State University of New Jersey, RutgersPiscataway, NJ, 08854, USA.
| |
Collapse
|
23
|
Korytina GF, Markelov VA, Gibadullin IA, Zulkarneev SR, Nasibullin TR, Zulkarneev RH, Avzaletdinov AM, Avdeev SN, Zagidullin NS. The Relationship Between Differential Expression of Non-coding RNAs (TP53TG1, LINC00342, MALAT1, DNM3OS, miR-126-3p, miR-200a-3p, miR-18a-5p) and Protein-Coding Genes (PTEN, FOXO3) and Risk of Idiopathic Pulmonary Fibrosis. Biochem Genet 2025:10.1007/s10528-024-11012-z. [PMID: 39881079 DOI: 10.1007/s10528-024-11012-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 12/20/2024] [Indexed: 01/31/2025]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a rapidly progressive interstitial lung disease of unknown pathogenesis with no effective treatment currently available. Given the regulatory roles of lncRNAs (TP53TG1, LINC00342, H19, MALAT1, DNM3OS, MEG3), miRNAs (miR-218-5p, miR-126-3p, miR-200a-3p, miR-18a-5p, miR-29a-3p), and their target protein-coding genes (PTEN, TGFB2, FOXO3, KEAP1) in the TGF-β/SMAD3, Wnt/β-catenin, focal adhesion, and PI3K/AKT signaling pathways, we investigated the expression levels of selected genes in peripheral blood mononuclear cells (PBMCs) and lung tissue from patients with IPF. Lung tissue and blood samples were collected from 33 newly diagnosed, treatment-naive patients and 70 healthy controls. Gene expression levels were analyzed by RT-qPCR. TaqMan assays and TaqMan MicroRNA assay were employed to quantify the expression of target lncRNAs, mRNAs, and miRNAs. Our study identified significant differential expression in PBMCs from IPF patients compared to healthy controls, including lncRNAs MALAT1 (Fold Change = 3.809, P = 0.0001), TP53TG1 (Fold Change = 0.4261, P = 0.0021), and LINC00342 (Fold Change = 1.837, P = 0.0448); miRNAs miR-126-3p (Fold Change = 0.102, P = 0.0028), miR-200a-3p (Fold Change = 0.442, P = 0.0055), and miR-18a-5p (Fold Change = 0.154, P = 0.0034); and mRNAs FOXO3 (Fold Change = 4.604, P = 0.0032) and PTEN (Fold Change = 2.22, P = 0.0011). In lung tissue from IPF patients, significant expression changes were observed in TP53TG1 (Fold Change = 0.2091, P = 0.0305) and DNM3OS (Fold Change = 4.759, P = 0.05). Combined analysis of PBMCs expression levels for TP53TG1, MALAT1, miRNA miR-126-3p, and PTEN distinguished IPF patients from healthy controls with an AUC = 0.971, sensitivity = 0.80, and specificity = 0.955 (P = 6 × 10-8). These findings suggest a potential involvement of the identified ncRNAs and mRNAs in IPF pathogenesis. However, additional functional validation studies are needed to elucidate the precise molecular mechanisms by which these lncRNAs, miRNAs, and their targets contribute to PF.
Collapse
Affiliation(s)
- Gulnaz F Korytina
- Institute of Biochemistry and Genetics-Subdivision of the Ufa Federal Research Centre of the Russian Academy of Sciences (IBG UFRC RAS), Pr. Oktyabrya, 71, Ufa, 450054, Russian Federation.
- Bashkir State Medical University, Lenina Str. 3, Ufa, 450008, Russian Federation.
| | - Vitaly A Markelov
- Institute of Biochemistry and Genetics-Subdivision of the Ufa Federal Research Centre of the Russian Academy of Sciences (IBG UFRC RAS), Pr. Oktyabrya, 71, Ufa, 450054, Russian Federation
- Bashkir State Medical University, Lenina Str. 3, Ufa, 450008, Russian Federation
| | - Irshat A Gibadullin
- Bashkir State Medical University, Lenina Str. 3, Ufa, 450008, Russian Federation
| | - Shamil R Zulkarneev
- Bashkir State Medical University, Lenina Str. 3, Ufa, 450008, Russian Federation
| | - Timur R Nasibullin
- Institute of Biochemistry and Genetics-Subdivision of the Ufa Federal Research Centre of the Russian Academy of Sciences (IBG UFRC RAS), Pr. Oktyabrya, 71, Ufa, 450054, Russian Federation
| | - Rustem H Zulkarneev
- Bashkir State Medical University, Lenina Str. 3, Ufa, 450008, Russian Federation
| | | | - Sergey N Avdeev
- Sechenov First Moscow State Medical University (Sechenov University), 8-2, Trubetskaya Str., Moscow, 119991, Russian Federation
| | - Naufal Sh Zagidullin
- Bashkir State Medical University, Lenina Str. 3, Ufa, 450008, Russian Federation
| |
Collapse
|
24
|
Jiang W, Wang P, Huang L. Upregulation of phosphatase and tensin homolog deleted on chromosome ten inhibits lung cancer cell proliferation by suppressing the oncogene polo-like kinase 1 and inducing autophagy. Cytojournal 2025; 22:10. [PMID: 39958887 PMCID: PMC11829310 DOI: 10.25259/cytojournal_146_2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 12/13/2024] [Indexed: 02/18/2025] Open
Abstract
Objective Lung cancer is one of the main causes of cancer-related mortality globally, and it poses considerable therapeutic challenges. Polo-like kinase 1 (PLK1) exhibits upregulation in lung cancer, and PLK1 silencing promotes autophagy in lung cancer cells, which inhibits tumor progression. The phosphatase and tensin homolog deleted on chromosome ten (PTEN) acts as a tumor suppressor gene. This study aimed to investigate whether PTEN regulates autophagy and inhibits lung cancer-cell proliferation by suppressing PLK1. Material and Methods In this study, we evaluated cell proliferation by silencing or overexpressing PLK1 and PTEN in A549 cells through 5-ethynyl-2'-deoxyuridine labeling and cloning experiments. The autophagy levels were detected through transmission electron microscopy, real-time quantitative polymerase chain reaction, and Western blot. Finally, the results of in vitro experiment were further verified using an in vivo xenograft tumor animal model. Results The upregulation of PTEN suppressed PLK1 expression in lung cancer cells and reduced their proliferation rate. In addition, the overexpression of PTEN has been associated with the growth of lung cancer tumors. In parallel, the levels of autophagy of lung cancer cells rose in response to PTEN upregulation in vivo and in vitro. Conclusion This study revealed that PTEN promotes the autophagy of lung cancer cells and inhibits cell proliferation and tumor growth by suppressing PLK1 expression. This finding provides a new strategy for lung cancer treatment by utilizing the autophagy-regulating effect of PTEN to inhibit lung cancer growth by targeting PLK1.
Collapse
Affiliation(s)
- Weizhou Jiang
- Department of Pulmonary Disease, Weifang Traditional Chinese Medicine Hospital, Weifang, Shandong, China
| | - Pei Wang
- Department of Pulmonary Disease, Weifang Traditional Chinese Medicine Hospital, Weifang, Shandong, China
| | - Limin Huang
- Department of Oncology, Weifang Traditional Chinese Medicine Hospital, Weifang, Shandong, China
| |
Collapse
|
25
|
Zhu M, Zhang S, Tang J, Hou H, Wang L, Lin H, Zhang X, Jin M. Two Small Peptides from Buthus martensii Hydrolysates Exhibit Antitumor Activity Through Inhibition of TNF-α-Mediated Signal Transduction Pathways. Life (Basel) 2025; 15:105. [PMID: 39860044 PMCID: PMC11766664 DOI: 10.3390/life15010105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/08/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
The scorpion Buthus martensii Karsch is edible and has been an essential resource in traditional Chinese medicine for treating numerous diseases. In this study, two small peptides from B. martensii hydrolysates were examined to elucidate their potential against gastric cancer. The small peptides (AK and GK) were identified using the LC-QTOF-MS-based approach. In silico prediction of therapeutic targets, MGC-803 cells and transgenic zebrafish models, and immunoblotting experiments were used to reveal the molecular mechanism of action of the peptides. The peptides AK and GK competitively bound to the receptor to modulate the TNF/TNFR-signaling cascade and alter the tumor microenvironment. EGFR, TP53, MYC, PTEN, and STAT3 were also identified as major functional targets of the peptides. Mechanistically, AK and GK inactivated the TNF-α/EGFR/STAT3-signaling pathway, decreased c-myc protein expression levels, and upregulated p53 and PTEN expression, thereby preventing TNF-α-induced tumor growth. Our findings indicated that AK and GK played a pivotal role in offsetting the inflammatory stimuli that caused gastric cancer cell invasion and highlighted the use of B. martensii resources as functional products with health benefits.
Collapse
Affiliation(s)
- Mengshuang Zhu
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (M.Z.); (S.Z.); (J.T.); (H.H.); (L.W.); (H.L.)
| | - Shanshan Zhang
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (M.Z.); (S.Z.); (J.T.); (H.H.); (L.W.); (H.L.)
| | - Jiyang Tang
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (M.Z.); (S.Z.); (J.T.); (H.H.); (L.W.); (H.L.)
| | - Hairong Hou
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (M.Z.); (S.Z.); (J.T.); (H.H.); (L.W.); (H.L.)
| | - Lizhen Wang
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (M.Z.); (S.Z.); (J.T.); (H.H.); (L.W.); (H.L.)
| | - Houwen Lin
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (M.Z.); (S.Z.); (J.T.); (H.H.); (L.W.); (H.L.)
- Research Center for Marine Drugs, State Key Laboratory of Oncogenes and Related Genes, Department of Pharmacy, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Xuanming Zhang
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (M.Z.); (S.Z.); (J.T.); (H.H.); (L.W.); (H.L.)
| | - Meng Jin
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (M.Z.); (S.Z.); (J.T.); (H.H.); (L.W.); (H.L.)
| |
Collapse
|
26
|
Ashadul Sk M, K H, Matada GSP, Pal R, B V M, Mounika S, E H, M P V, D A. Current developments in PI3K-based anticancer agents: Designing strategies, biological activity, selectivity, structure-activity correlation, and docking insight. Bioorg Chem 2025; 154:108011. [PMID: 39662340 DOI: 10.1016/j.bioorg.2024.108011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 12/13/2024]
Abstract
The phospatidylinositol-3 kinase (PI3K) pathway is a critical intracellular signalling mechanism that is changed or amplified in a variety of cancers, including breast, gastric, ovarian, colorectal, prostate, glioma, and endometrial. PI3K signalling is important for cancer cell survival, angiogenesis, and metastasis, making it a promising therapeutic target. The PI3K kinases in their different isoforms, namely α, β, δ, and γ, encode PIK3CA, PIK3CB, PIK3CD, and PIK3CG genes. Specific gene mutation or overexpression of the protein is responsible for the therapeutic failure of current therapeutics. There are several current and completed clinical trials using PI3K inhibitors (pan, isoform-specific, and dual PI3K/mTOR) to develop effective PI3K inhibitors capable of overcoming resistance to existing drugs. However, the bulk of these inhibitors have had their indications revoked or voluntarily withdrawn due to concerns about their harmful consequences. Several inhibitors containing medicinally privileged scaffolds like thiazole, triazine, benzimidazole, podophyllotoxin, pyridine, quinazoline, thieno-triazole, pyrimidine, triazole, benzofuran, imidazo-pyridazine, oxazole, coumarin, and azepine derivatives have been explored to target the PI3K pathway and/or a specific isoform in the current overview. This article reviews the structure, biological activities, and clinical status of PI3K inhibitors. It focuses on the development techniques, docking insight, and structure-activity connections of PI3K-based inhibitors. The findings provide useful insights and future approaches for the development of promising PI3K-based inhibitors.
Collapse
Affiliation(s)
- Md Ashadul Sk
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru 560107, Karnataka, India
| | - Hemalatha K
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru 560107, Karnataka, India.
| | - Gurubasavaraja Swamy Purawarga Matada
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru 560107, Karnataka, India
| | - Rohit Pal
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru 560107, Karnataka, India.
| | - Manjushree B V
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru 560107, Karnataka, India
| | - S Mounika
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru 560107, Karnataka, India
| | - Haripriya E
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru 560107, Karnataka, India
| | - Viji M P
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru 560107, Karnataka, India
| | - Anjan D
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru 560107, Karnataka, India
| |
Collapse
|
27
|
Yang J, Yang H, Wang F, Dai Y, Deng Y, Shi K, Zhu Z, Liu X, Ma X, Gao Y. Bioinformatics identification based on causal association inference using multi-omics reveals the underlying mechanism of Gui-Zhi-Shao-Yao-Zhi-Mu decoction in modulating rheumatoid arthritis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156332. [PMID: 39736250 DOI: 10.1016/j.phymed.2024.156332] [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: 06/02/2024] [Revised: 12/08/2024] [Accepted: 12/15/2024] [Indexed: 01/01/2025]
Abstract
OBJECT Rheumatoid arthritis (RA) is a prevalent and currently incurable autoimmune disease. Existing conventional medical treatments are limited in their efficacy, prolonged disease may lead to bone destruction, joint deformity, and loss of related functions, which places a huge burden on RA patients and their families. For millennia, the use of traditional Chinese medicine (TCM), exemplified by the Gui-Zhi-Shao-Yao-Zhi-Mu decoction (GZSYZM), has been demonstrated to offer distinct therapeutic advantages in the management of RA. Exploring the potential mechanism of GZSYZM in the treatment of RA is a hot topic in the field of TCM. METHOD High-throughput sequencing data of RA at bulk level and single cell level and Chinese Materia Medica target-related databases were used as data sources. Ultra-high-performance liquid chromatography coupled with high-resolution mass spectrometry was employed for the identification of the most relevant compounds to the active ingredients present in the GZSYZM granules. Potential disease genes were identified using a combination of differential expression analysis and weighted gene co-expression network analysis, and the "Chinese Materia Medica-Ingredient-Target" network was constructed to obtain candidate drug target genes. The GZSYZM-RA hub genes were then identified based on Molecular Complex Detection algorithm. To explore the associations and potential mechanisms between the GZSYZM-RA hub gene set and RA, Mendelian randomization (MR) analysis and Bayesian co-localization analysis were used to further identify the GZSYZM-RA core genes that were causally associated with RA. A nomogram was constructed based on a multifactorial logistic regression model using the GZSYZM-RA core genes as predictors of RA. To evaluate its diagnostic value, receiver operating characteristic (ROC) curves, calibration curves, and decision curves were plotted. The potential downstream regulatory mechanisms of the gene of interest in GZSYZM in RA therapy were finally investigated using single- gene set enrichment analysis and molecular docking. The aim was to model the optimal conformation of its target protein receptor binding to the small molecule ligand in GZSYZM to identify the key constituents. RESULT Functional enrichment analysis revealed that the GZSYZM-RA hub gene set is enriched in several autoimmune-related mechanistic pathways, with a particular emphasis on the phosphoinositide 3 kinase (PI3K)‑serine/threonine kinase (AKT) signaling pathway. AUCell scores demonstrated active expression of the GZSYZM-RA hub gene set with the PI3K-AKT signaling pathway on monocytes, especially non-classical monocytes. Immunol infiltration analysis based on the CIBERSORT algorithm also showed a strong correlation between several genes in the GZSYZM-RA hub gene set and monocytes by calculating Spearman's rank correlation coefficients. MR analysis with co-localization analysis further identified seven core genes (CASP8, PPARG, IKBKB, PPARA, IFNG, MYC, and STAT3) causally associated with RA. Diagnostic value for clinical decision making was demonstrated by a multivariable logistic regression model constructed with GZSYZM-RA core genes. Molecular docking analysis indicates that CASP8 and GZSYZM have high docking scores, with three key constituents (quercetin, kaempferol, and diosmetin) exhibiting strong binding affinities. CONCLUSION GZSYZM may regulate the abnormal over-proliferation and apoptotic imbalance of fibroblast-like synoviocytes in RA patients by inhibiting signaling of the PI3K-AKT signaling pathway while activating CASP8-mediated pro-apoptotic effects. And it may be effective in directly or indirectly inhibiting monocyte-to-osteoclast differentiation, ultimately improving the poor prognosis of joint destruction in RA patients.
Collapse
Affiliation(s)
- Jiayue Yang
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Heng Yang
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Fumin Wang
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Yao Dai
- The Fourth Clinical Medical College of Xinjiang Medical University, The Fourth Affiliated Hospital of Xinjiang Medical University, Xinjiang Medical University Graduate School, Xinjiang Medical University, Xinjiang Uygur Autonomous Region 830054, China; Chinese Medicine Hospital of Gao County, Yibin 645150, China
| | - Yuxuan Deng
- Eye School of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Kaiyun Shi
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, No. 16 Jichang Road, Baiyun District, Guangzhou 510405, China
| | - Zehua Zhu
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China; Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Xinkun Liu
- Department of Acupuncture, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Yongxiang Gao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China.
| |
Collapse
|
28
|
Wei L, Liang Q, Zhou C, Liu R, Liu Y. PTEN inhibits epithelial mesenchymal transition of thyroid cancer cells by regulating the Wnt/β-Catenin signaling pathway. Discov Oncol 2024; 15:803. [PMID: 39692895 DOI: 10.1007/s12672-024-01596-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 11/15/2024] [Indexed: 12/19/2024] Open
Abstract
OBJECTIVE The global incidence of thyroid cancer (THCA) has significantly risen in recent years. This study aims to investigate the role and mechanisms of PTEN in epithelial mesenchymal transition (EMT), invasion and migration of THCA cells. METHODS PTEN expression in THCA was analyzed through bioinformatics databases. RT-qPCR and Western blot analyses were performed to quantify PTEN levels in the Nthy-ori 3-1 cell line and three THCA cell types (TPC-1, B-CPAP, FTC-133). TPC-1 cells were transfected with a PTEN overexpression plasmid and treated with the Wnt activator. Cell viability and apoptosis were assessed via CCK-8 and flow cytometry, respectively. The expression levels of E-Cadherin, N-Cadherin, and Vimentin in TPC-1 cells were evaluated using Western blot. The invasive, migratory, and wound-healing abilities of the cells were examined using Transwell and scratch assays. Activation of the Wnt/β-catenin pathway was assessed through Western blot. RESULTS PTEN expression was significantly lower in THCA cells, particularly in TPC-1 cells compared to other cell lines. PTEN overexpression led to decreased viability in TPC-1 cells, increased apoptosis, and a rise in E-Cadherin levels while reducing N-Cadherin and Vimentin levels, thereby inhibiting EMT. Furthermore, PTEN overexpression diminished the invasive, migratory and wound-healing capabilities of TPC-1 cells and suppressed activation of the Wnt/β-catenin pathway. Treatment with the Wnt activator partially counteracted the effects of PTEN overexpression on TPC-1 cells. CONCLUSION PTEN functions to inhibit EMT and the invasive and migratory characteristics of THCA cells by blocking the activation of the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Li Wei
- Department of Ultrasound, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, 443000, Hubei, China.
| | - Qianhui Liang
- Department of Oncology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, 443000, Hubei, China
| | - Chang Zhou
- Department of Ultrasound, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, 443000, Hubei, China
| | - Rong Liu
- Department of Ultrasound, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, 443000, Hubei, China
| | - Yun Liu
- Department of Ultrasound, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, 443000, Hubei, China
| |
Collapse
|
29
|
Nelson AB, Reese LE, Rono E, Queathem ED, Qiu Y, McCluskey BM, Crampton A, Conniff E, Cummins K, Boytim E, Dansou S, Hwang J, Safo S, Puchalska P, Wood DK, Schwertfeger KL, Crawford PA. Deciphering Colorectal Cancer-Hepatocyte Interactions: A Multiomic Platform for Interrogation of Metabolic Crosstalk in the Liver-Tumor Microenvironment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.06.627264. [PMID: 39713297 PMCID: PMC11661097 DOI: 10.1101/2024.12.06.627264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Metabolic reprogramming is a hallmark of cancer, enabling tumor cells to adapt to and exploit their microenvironment for sustained growth. The liver is a common site of metastasis, but the interactions between tumor cells and hepatocytes remain poorly understood. In the context of liver metastasis, these interactions play a crucial role in promoting tumor survival and progression. This study leverages multiomics coverage of the microenvironment via liquid chromatography and high-resolution, high-mass accuracy mass spectrometry-based untargeted metabolomics, 13C-stable isotope tracing, and RNA sequencing to uncover the metabolic impact of co-localized primary hepatocytes and a colon adenocarcinoma cell line, SW480, using a 2D co-culture model. Metabolic profiling revealed disrupted Warburg metabolism with an 80% decrease in glucose consumption and 94% decrease in lactate production by hepatocyte-SW480 co-cultures relative to SW480 control cultures. Decreased glucose consumption was coupled with alterations in glutamine and ketone body metabolism, suggesting a possible fuel switch upon co-culturing. Further, integrated multiomic analysis indicates that disruptions in metabolic pathways, including nucleoside biosynthesis, amino acids, and TCA cycle, correlate with altered SW480 transcriptional profiles and highlight the importance of redox homeostasis in tumor adaptation. Finally, these findings were replicated in 3-dimensional microtissue organoids. Taken together, these studies support a bioinformatic approach to study metabolic crosstalk and discovery of potential therapeutic targets in preclinical models of the tumor microenvironment.
Collapse
Affiliation(s)
- Alisa B. Nelson
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN USA
| | - Lyndsay E. Reese
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN USA
| | - Elizabeth Rono
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN USA
| | - Eric D. Queathem
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN USA
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN USA
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Yinjie Qiu
- Minnesota Supercomputing Institute, Minneapolis, MN USA
| | | | - Alexandra Crampton
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN USA
| | - Eric Conniff
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN USA
| | - Katherine Cummins
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN USA
| | - Ella Boytim
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN USA
| | - Senali Dansou
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN USA
| | - Justin Hwang
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN USA
| | - Sandra Safo
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, MN, USA
| | - Patrycja Puchalska
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN USA
| | - David K. Wood
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN USA
| | - Kathryn L. Schwertfeger
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN USA
| | - Peter A. Crawford
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN USA
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN USA
| |
Collapse
|
30
|
Timofeeva AV, Fedorov IS, Nikonets AD, Tarasova AM, Balashova EN, Degtyarev DN, Sukhikh GT. Increased Levels of hsa-miR-199a-3p and hsa-miR-382-5p in Maternal and Neonatal Blood Plasma in the Case of Placenta Accreta Spectrum. Int J Mol Sci 2024; 25:13309. [PMID: 39769074 PMCID: PMC11678653 DOI: 10.3390/ijms252413309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/08/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Despite the increasing number of placenta accreta spectrum (PAS) cases in recent years, its impact on neonatal outcomes and respiratory morbidity, as well as the underlying pathogenetic mechanism, has not yet been extensively studied. Moreover, no study has yet demonstrated the effectiveness of antenatal corticosteroid therapy (CT) for the prevention of respiratory distress syndrome (RDS) in newborns of mothers with PAS at the molecular level. In this regard, microRNA (miRNA) profiling by small RNA deep sequencing and quantitative real-time PCR was performed on 160 blood plasma samples from preterm infants (gestational age: 33-36 weeks) and their mothers who had been diagnosed with or without PAS depending on the timing of the antenatal RDS prophylaxis. A significant increase in hsa-miR-199a-3p and hsa-miR-382-5p levels was observed in the blood plasma of the newborns from mothers with PAS compared to the control group. A clear trend toward the normalization of hsa-miR-199a-3p and hsa-miR-382-5p levels in the neonatal blood plasma of the PAS groups was observed when CT was administered within 14 days before delivery, but not beyond 14 days. Direct correlations were found among the hsa-miR-382-5p level in neonatal blood plasma and the hsa-miR-199a-3p level in the same sample (r = 0.49; p < 0.001), the oxygen requirements in the NICU (r = 0.41; p = 0.001), the duration of the NICU stay (r = 0.31; p = 0.019), and the severity of the newborn's condition based on the NEOMOD scale (r = 0.36; p = 0.005). Logistic regression models based on the maternal plasma levels of hsa-miR-199a-3p and hsa-miR-382-5p predicted the need for cardiotonic therapy, invasive mechanical ventilation, or high-frequency oscillatory ventilation in newborns during the early neonatal period, with a sensitivity of 95-100%. According to the literary data, these miRNAs regulate fetal organogenesis via IGF-1, the formation of proper lung tissue architecture, surfactant synthesis in alveolar cells, and vascular tone.
Collapse
Affiliation(s)
- Angelika V. Timofeeva
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician Kulakov V.I., 117997 Moscow, Russia
| | | | | | | | | | | | | |
Collapse
|
31
|
Mi J, Zhao L, Shen Y, Mo S, Kuang Y. PFKP Lactylation Promotes the Ovarian Cancer Progression Through Targeting PTEN. Biochem Genet 2024:10.1007/s10528-024-10990-4. [PMID: 39638933 DOI: 10.1007/s10528-024-10990-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024]
Abstract
Ovarian cancer (OC) ranks among the most prevalent malignancies affecting females globally and is a leading cause of cancer-related mortality in women. This study sought to elucidate the influence of phosphofructokinase P (PFKP) on the progression of OC. A cohort of sixty OC patients was enrolled. OC cells were exposed to both normoxic and hypoxic conditions. Expression levels of PFKP and phosphatase and tensin homolog (PTEN) were quantified using real time quantitative polymerase chain reaction (RT-qPCR) and Western blot analyses. Immunofluorescence confirmed these protein expression patterns. Glycolysis-related parameters, encompassing glucose uptake, extracellular lactate levels, extracellular acidification rates, and oxygen consumption rates, were assessed using commercially available kits. Lactylation status of PFKP was evaluated via immunoprecipitation followed by Western blot analysis. An OC xenograft mouse model was also established. Findings indicated elevated PFKP expression in OC tissues and cells. Additionally, PFKP knockdown attenuated glycolysis and counteracted the hypoxia-induced enhancement of glycolytic activity in OC cells. Mutation of the lysine (K) residue at position 392 diminished PFKP lactylation. Further investigations revealed that PFKP depletion upregulated PTEN expression in hypoxia-treated OC cells. Besides, PTEN suppression increased the glycolysis in hypoxia-treated OC cells. Animal study results demonstrated that PFKP inhibition curtailed OC tumor growth by modulating PTEN expression. Collectively, these results suggested that lactylation of PFKP at the K392 residue promoted glycolysis in OC cells by regulating PTEN, thereby facilitating the disease's progression.
Collapse
Affiliation(s)
- Jianfeng Mi
- Department of Gynecology, The First Affiliated Hospital of Guangxi Medical University, Nanning, No.6 Shaungyong Road, Nanning, 532200, China
| | - Ling Zhao
- Department of Gynecology, The First Affiliated Hospital of Guangxi Medical University, Nanning, No.6 Shaungyong Road, Nanning, 532200, China
| | - Yonglong Shen
- Department of Gynecology, The First Affiliated Hospital of Guangxi Medical University, Nanning, No.6 Shaungyong Road, Nanning, 532200, China
| | - Shien Mo
- Department of Gynecology, The Sec People's Hospital of Qinzhou, Qinzhou, China
| | - Yan Kuang
- Department of Gynecology, The First Affiliated Hospital of Guangxi Medical University, Nanning, No.6 Shaungyong Road, Nanning, 532200, China.
| |
Collapse
|
32
|
Wang R, Yu H, Li A, Wang T, Wang Q, Qi H, Wang C, Zhang J. Maslinic acid prevented lipopolysaccharide-induced injury of IPEC-J2 cells through regulating PTEN-FAK signaling pathway. J Cell Physiol 2024; 239:e31446. [PMID: 39311471 DOI: 10.1002/jcp.31446] [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: 03/15/2024] [Revised: 09/05/2024] [Accepted: 09/13/2024] [Indexed: 12/18/2024]
Abstract
Intestinal epithelial injury is one of the typical symptoms associated with intestinal inflammation and diarrhea, and the repair of the intestinal epithelium intricately linked to cell migration. Here, we test the hypothesis that maslinic acid (MA) regulates porcine intestinal epithelial cell migration by inhibiting focal adhesion kinase (FAK)/AKT signaling pathway. In this experiment, the optimal concentration of MA (0.5 μg/mL) on IPEC-J2 cell viability was selected to investigate the effect under low-dose lipopolysaccharide (LPS) (1 μg/mL) conditions. Transcriptome sequencing and polymerase chain reaction array results revealed that MA could alleviate LPS-induced the gene expressions decreasing in focal adhesion signaling pathway. From the pathway map analysis and western blot analysis results, MA alleviated the LPS-induced decrease in FAK protein expression mainly by promoting FAK protein phosphorylation, which in turn alleviated the decrease in cell migration and formation of cytoskeleton protein Vinculin and F-actin, the above results were verified by FAK phosphorylation inhibitors Defactinib. The molecular docking and immunoprecipitation further verified that MA could bind to PTEN protein and significantly inhibit its interaction with FAK protein, blocking the function of PTEN to inhibit FAK phosphorylation finally shown to promote the level of FAK phosphorylation, meanwhile LPS inhibited FAK protein expression and its binding to PKC and PTEN proteins. Our study revealed the role of MA and LPS in FAK protein, and increased understanding of MA anti-inflammatory mechanism.
Collapse
Affiliation(s)
- Rui Wang
- College of Animal Sciences, Jilin University, Changchun, China
| | - Hao Yu
- College of Animal Sciences, Jilin University, Changchun, China
| | - Aike Li
- Academy of National Food and Strategic Reserves Administration, Beijing, China
| | - Ting Wang
- College of Animal Sciences, Jilin University, Changchun, China
| | - Qiyuan Wang
- College of Animal Sciences, Jilin University, Changchun, China
| | - Huiyu Qi
- College of Animal Sciences, Jilin University, Changchun, China
| | - Chuanqi Wang
- College of Animal Sciences, Jilin University, Changchun, China
| | - Jing Zhang
- College of Animal Sciences, Jilin University, Changchun, China
- Academy of National Food and Strategic Reserves Administration, Beijing, China
| |
Collapse
|
33
|
Dai Z, Zhao W, Cao L, Zhu Z, Xia Z, Xia L. Engineered probiotic E.coli Nissle 1917 for release PTEN to improve the tumor microenvironment and suppress tumor growth. Biotechnol Lett 2024; 46:1237-1247. [PMID: 39331305 DOI: 10.1007/s10529-024-03536-6] [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: 07/05/2024] [Revised: 09/05/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024]
Abstract
The cancer is one of the diseases of serious threat to people's health and life nowadays. But heterogeneity, drug resistance and treatment side effects of cancer, traditional treatments still have limitations. Tumor-targeting probiotics with a well-established Biosafety and efficient targeting as a delivery vectors to deliver anticancer genes or antitumor drugs to tumor microenvironment has attracted much attention in cancer therapies. In this study, E.coil Nissle 1917 (EcN) was utilized to deliver eukaryotic anti-tumor protein PTEN to tumor microenvironment and suppress tumor growth. Therefore, the EcN (PTEN) was developed. Our results demonstrated that EcN (PTEN) could colonize the tumor site accurately and inhibit the growth of colorectal cancer cells in tumor-bearing mice. It is worth noting that the tumor microenvironment of the treated mice showed significant recruitment of and M1 macrophages, neutrophils and T lymphocytes. No toxicity was observed in the normal tissues during the experiments. This research show the probiotic EcN(PTEN) holds the promise of becoming a powerful weapon against cancer and expected to provide more effective treatments for cancer patients.
Collapse
Affiliation(s)
- Zirui Dai
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Provincial Key Laboratory of Microbial Molecular Biology, College of Life Science, Hunan Normal University, No. 36 Lushan Street, Changsha, 410081, People's Republic of China
| | - Wenjuan Zhao
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Provincial Key Laboratory of Microbial Molecular Biology, College of Life Science, Hunan Normal University, No. 36 Lushan Street, Changsha, 410081, People's Republic of China
| | - Li Cao
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Provincial Key Laboratory of Microbial Molecular Biology, College of Life Science, Hunan Normal University, No. 36 Lushan Street, Changsha, 410081, People's Republic of China
| | - Zirong Zhu
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Provincial Key Laboratory of Microbial Molecular Biology, College of Life Science, Hunan Normal University, No. 36 Lushan Street, Changsha, 410081, People's Republic of China
| | - Ziyuan Xia
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Provincial Key Laboratory of Microbial Molecular Biology, College of Life Science, Hunan Normal University, No. 36 Lushan Street, Changsha, 410081, People's Republic of China
| | - Liqiu Xia
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Provincial Key Laboratory of Microbial Molecular Biology, College of Life Science, Hunan Normal University, No. 36 Lushan Street, Changsha, 410081, People's Republic of China.
| |
Collapse
|
34
|
Farinha D, Sarmento-Ribeiro AB, Faneca H. Combination of Gene Therapy and Chemotherapy in a New Targeted Hybrid Nanosystem to Hepatocellular Carcinoma. Int J Nanomedicine 2024; 19:12505-12527. [PMID: 39606562 PMCID: PMC11598603 DOI: 10.2147/ijn.s474665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024] Open
Abstract
Purpose Hepatocellular carcinoma is the most frequent liver cancer and constitutes one of the main causes of cancer mortality. The combination of targeted therapy drugs, such as selumetinib and perifosine that inhibit cell signaling pathways involved in cell survival and proliferation, with the expression of tumor suppressor transgenes, such as PTEN, may result in an efficient therapeutic approach against HCC. Thus, the main objective of this work was to develop a new lipid-polymer hybrid nanosystem (HNP), composed of a PLGA core coated with a pH-sensitive lipid bilayer functionalized with the targeting ligand GalNAc, in order to specifically and efficiently deliver this novel combination of therapeutic agents in HCC cells. Methods Transmission electron microscopy, zeta potential, Fourier transform infrared spectroscopy, and dynamic light scattering were used to determine the physicochemical properties of hybrid nanosystems and their components. The biological activity and specificity of nanosystems were evaluated using luminescence and flow cytometry. A variety of techniques were used to assess the therapeutic activity of hybrid nanosystems, including the Alamar Blue assay for cell viability; flow cytometry for cell death mechanisms, mitochondrial membrane potential and cell cycle; luminescence for caspase activity; flow cytometry and fluorescence microscopy for cell proliferation; and Western blot for molecular targets levels. Results The obtained results showed that this new hybrid nanosystem not only has a high loading capacity of both drugs, but also allows for substantial expression of the PTEN transgene. In addition, the developed formulation has high stability, adequate physicochemical properties and high specificity to HCC cells. Moreover, the achieved data revealed that this innovative nanosystem presents a high antitumor effect, demonstrated not only by the enhancement on the programmed cell death, but also by the reduction in cell proliferation capacity. Conclusion The generated formulation shows a high anticancer effect, demonstrating a high translational potential for future clinical application in HCC treatment.
Collapse
Affiliation(s)
- Dina Farinha
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Ana Bela Sarmento-Ribeiro
- CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Laboratory of Oncobiology and Hematology (LOH) and University Clinic of Hematology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Clinical Hematology Department, Centro Hospitalar Universitário de Coimbra (CHUC), Coimbra, Portugal
| | - Henrique Faneca
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
35
|
Xie AX, Tansey W, Reznik E. UnitedMet harnesses RNA-metabolite covariation to impute metabolite levels in clinical samples. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.24.24307903. [PMID: 38826234 PMCID: PMC11142294 DOI: 10.1101/2024.05.24.24307903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Comprehensively studying metabolism requires the measurement of metabolite levels. However, in contrast to the broad availability of gene expression data, metabolites are rarely measured in large molecularly-defined cohorts of tissue samples. To address this basic barrier to metabolic discovery, we propose a Bayesian framework ("UnitedMet") which leverages the empirical strength of RNA-metabolite covariation to impute otherwise unmeasured metabolite levels from widely available transcriptomic data. We demonstrate that UnitedMet is equally capable of imputing whole pool sizes as well as the outcomes of isotope tracing experiments. We apply UnitedMet to investigate the metabolic impact of driver mutations in kidney cancer, identifying a novel association between BAP1 and a highly oxidative tumor phenotype. We similarly apply UnitedMet to determine that advanced kidney cancers upregulate oxidative phosphorylation relative to early-stage disease, that oxidative metabolism in kidney cancer is associated with inferior outcomes to combination therapy, and that kidney cancer metastases themselves demonstrate elevated oxidative phosphorylation relative to primary tumors. UnitedMet therefore enables the assessment of metabolic phenotypes in contexts where metabolite measurements were not taken or are otherwise infeasible, opening new avenues for the generation and evaluation of metabolite-centered hypotheses. UnitedMet is open source and publicly available (https://github.com/reznik-lab/UnitedMet).
Collapse
Affiliation(s)
- Amy X. Xie
- Computational Oncology Service, Memorial Sloan Kettering Cancer Center, NY NY 10065
- Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY
| | - Wesley Tansey
- Computational Oncology Service, Memorial Sloan Kettering Cancer Center, NY NY 10065
| | - Ed Reznik
- Computational Oncology Service, Memorial Sloan Kettering Cancer Center, NY NY 10065
| |
Collapse
|
36
|
You H, Gou Q, Dong M, Chang F, Xiu J. Exploring the role of iNOS in HFpEF-Related myocardial fibrosis: Involvement of PTEN-PI3K/AKT signaling pathway. Biochem Biophys Res Commun 2024; 734:150589. [PMID: 39245028 DOI: 10.1016/j.bbrc.2024.150589] [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: 07/04/2024] [Revised: 08/06/2024] [Accepted: 08/21/2024] [Indexed: 09/10/2024]
Abstract
BACKGROUND Heart failure with preserved ejection fraction (HFpEF) is a challenging condition to treat with myocardial fibrosis being a pivotal pathological component. Previous studies have suggested a role for inducible nitric oxide synthase (iNOS) in the progression of this condition, but the precise mechanisms remain unclear. This study aimed to investigate the role of iNOS in HFpEF-related myocardial fibrosis and identify potential therapeutic targets. METHODS A 'two-hit' mouse model of HFpEF was established, and echocardiography, histopathology and biochemical analyses were performed. In vitro experiments were conducted in mouse cardiac fibroblasts, with iNOS overexpression and application of iNOS or phosphatidylinositol 3 kinase (PI3K) inhibitors. The iNOS-S-nitrosylated phosphatase and TENsin homolog (SNO-PTEN)-phosphorylated-protein kinase B (p-AKT) pathway was investigated, along with the effects on fibrotic markers and cell proliferation and migration. RESULTS HFpEF mice exhibited significant cardiac dysfunction and fibrosis, with increased expression of iNOS, SNO-PTEN, and p-AKT, indicative of the activation of the iNOS-SNO-PTEN-p-AKT pathway. iNOS overexpression in mouse cardiac fibroblasts led to increased SNO-PTEN, decreased PTEN, activated phosphorylated PI3K (p-PI3K) and p-AKT, and enhanced cell proliferation and migration, as well as increased collagen I and III expression. The use of an iNOS inhibitor (L-NIL) or a PI3K inhibitor (LY294002) partially reversed these changes. CONCLUSION Our findings suggest that the iNOS-SNO-PTEN-p-AKT pathway may play a crucial role in HFpEF-related myocardial fibrosis, with iNOS and PI3K inhibitors offering potential therapeutic benefits. These insights may pave the way for the development of effective drug therapies for HFpEF.
Collapse
Affiliation(s)
- Hongjun You
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China; Department of Cardiovascular Medicine, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
| | - Qiling Gou
- Department of Cardiovascular Medicine, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
| | - Mengya Dong
- Department of Cardiovascular Medicine, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
| | - Fengjun Chang
- Department of Cardiovascular Medicine, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
| | - Jiancheng Xiu
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
37
|
Sánchez-Castillo A, Kampen KR. Understanding serine and glycine metabolism in cancer: a path towards precision medicine to improve patient's outcomes. Discov Oncol 2024; 15:652. [PMID: 39538085 PMCID: PMC11561223 DOI: 10.1007/s12672-024-01544-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024] Open
Abstract
In this perspective, we highlight and reflect on the current knowledge with respect to serine/glycine metabolism in cancer, therapeutic resistance, and precision medicine opportunities for therapeutic targeting and treatment follow-up. Cancer subtypes with high mortality rates include lung cancer and glioblastomas. In order to improve future therapeutic opportunities, patient stratification need to be performed to select patients that might benefit from adjuvant serine/glycine targeting compounds. In an effort to identify the group of patients for stratification purposes, we analyzed publicly available TCGA patient datasets to test associations between serine/glycine metabolism enzyme expression and important cancer drivers in lung cancer and glioblastoma. These patients presenting serine/glycine pathway overexpression might benefit from adjuvant sertraline treatment in the future.
Collapse
Affiliation(s)
- Anaís Sánchez-Castillo
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht University, Maastricht, The Netherlands
| | - Kim R Kampen
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht University, Maastricht, The Netherlands.
- Department of Oncology, Laboratory for Disease Mechanisms in Cancer, KU Leuven and Leuven Cancer Institute (LKI), Louvain, Belgium.
| |
Collapse
|
38
|
Ren LL, Song YR, Song ZC, Yang H, Zhang Q, Ji MM, Xiao N, Wen M, Wang JH. Enhancing antitumor activity of herceptin in HER2-positive breast cancer cells: a novel DNMT-1 inhibitor approach. Discov Oncol 2024; 15:640. [PMID: 39527385 PMCID: PMC11555163 DOI: 10.1007/s12672-024-01508-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: 07/23/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
HER2 antagonists remain the cornerstone of therapy for patients with HER2-positive breast cancer. This study introduces a novel small-molecule inhibitor of DNA methyltransferase 1 (DNMT-1), referred to as DI-1, designed to synergize with HER2 antagonists in treating HER2-positive breast cancer cells. Clinical data reveal a negative correlation between DNMT-1 expression and PTEN levels, and a positive correlation with the methylation rates of PTEN's promoter. In experiments with SKBR3 and BT474 cells, DI-1 effectively reduced the methylation of PTEN's promoter region, thereby upregulating PTEN expression. This upregulation, in turn, enhanced the cells' sensitivity to HER2 antagonists, indicating that DI-1's mechanism involves inhibiting DNMT-1's recruitment to PTEN's promoter region. Consequently, by increasing PTEN expression, DI-1 amplifies the sensitivity of HER2-positive breast cancer cells to treatment, suggesting its potential as a promising therapeutic strategy in this context.
Collapse
Affiliation(s)
- Li-Li Ren
- Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding, 071000, Hebei Province, People's Republic of China
| | - Yan-Ru Song
- Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding, 071000, Hebei Province, People's Republic of China
| | - Zhen-Chuan Song
- Department of Breast Center, Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei Province, People's Republic of China
| | - Hua Yang
- Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding, 071000, Hebei Province, People's Republic of China.
| | - Qian Zhang
- Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding, 071000, Hebei Province, People's Republic of China
| | - Meng-Meng Ji
- Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding, 071000, Hebei Province, People's Republic of China
| | - Na Xiao
- Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding, 071000, Hebei Province, People's Republic of China
| | - Ming Wen
- Department of Surgery, the Affiliated Hospital of Hebei University, Baoding, 071000, Hebei Province, People's Republic of China
| | - Ji-Hai Wang
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou City, 450052, Henan Province, People's Republic of China.
| |
Collapse
|
39
|
Hazazi A, Khan FR, Albloui F, Arif S, Abdulaziz O, Alhomrani M, Sindi AAA, Abu-Alghayth MH, Abalkhail A, Nassar SA, Binshaya AS. Signaling pathways in HPV-induced cervical cancer: Exploring the therapeutic promise of RNA modulation. Pathol Res Pract 2024; 263:155612. [PMID: 39357186 DOI: 10.1016/j.prp.2024.155612] [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: 07/31/2024] [Revised: 09/05/2024] [Accepted: 09/24/2024] [Indexed: 10/04/2024]
Abstract
Cervical cancer, originating from the epithelial tissue of the uterine cervix, constitutes the most commonly diagnosed malignancy among women worldwide. The predominant etiological factor underpinning cervical carcinogenesis is persistent infection with high-risk human papillomavirus (HPV) genotypes, notably HPV-16 and HPV-18. Oncoproteins encoded by high-risk HPV interfere with multiple essential cellular signaling cascades. Specifically, E5, E6, and E7 proteins disrupt the signaling pathways like p53, retinoblastoma tumor suppressor protein (pRB), The phosphoinositide 3 kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR), epidermal growth factor receptor (EGFR), mitogen-activated protein kinases (MAPK)/extracellular signal-regulated kinases (ERK), and Wnt/β-catenin, promoting HPV-mediated carcinogenesis. This dysregulation disrupts cell cycle control, apoptosis, and metastasis through modulation of microRNAs (miRNA) and key cellular processes. The novel therapeutic interventions for HPV prevention and detection are fundamental to patient management. RNA-based treatment modalities offer the potential for manipulating critical pathways involved in cervical carcinogenesis. RNA therapeutics offer novel approaches to drug development by targeting intracellular genetic elements inaccessible to conventional modalities. Additional advantages include rapid design, synthesis, and a reduced genotoxic profile compared to DNA-based therapies. Despite beneficial attributes, system stability and efficient delivery remain critical parameters. This study assessed the intricate relationship between HPV, cervical cancer, and various signaling pathways. The study explores miRNAs' diagnostic and therapeutic potential, mall interfering RNAs (siRNAs), and long non-coding RNAs (lncRNAs)in cervical cancer management. The review highlights the prospect of RNA-targeted therapies to modulate specific cancer signaling pathways. This approach offers a novel strategy for cervical cancer treatment through precise regulation of cancer signaling. Future research should concentrate on developing RNA-targeted interventions to improve cervical cancer treatment outcomes through increased therapeutic efficacy and specificity.
Collapse
Affiliation(s)
- Ali Hazazi
- Department of Pathology and Laboratory Medicine, Security Forces Hospital Program, Riyadh, Saudi Arabia; College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Farhan R Khan
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, Al Quwayiyah, Shaqra University, Riyadh, Saudi Arabia; Department of Pharmaceutical Chemistry, Azad Institute of Pharmacy and Research, Lucknow, UP, India
| | - Fawaz Albloui
- Department of Pathology and Laboratory Medicine, Security Forces Hospital Program, Riyadh, Saudi Arabia
| | - Sultan Arif
- Department of Plastic Surgery and Burn Unit, Security Force Hospital, Riyadh, Saudi Arabia
| | - Osama Abdulaziz
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O.Box 11099, Taif 21944, Saudi Arabia
| | - Majid Alhomrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O.Box 11099, Taif 21944, Saudi Arabia; Research Centre for Health Sciences, Taif University, Taif, Saudi Arabia
| | - Abdulmajeed A A Sindi
- Department of Basic Medical Sciences, Faculty of Applied Medical Sciences, Al-Baha University, Al-Baha, Saudi Arabia
| | - Mohammed H Abu-Alghayth
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, P.O. Box 255, Bisha 67714, Saudi Arabia
| | - Adil Abalkhail
- Department of Public Health, College of Applied Medical Sciences, Qassim University, Buraydah, P.O.Box 66666, Saudi Arabia
| | - Somia A Nassar
- Department of Medical Laboratory Science, College of Applied Medical Sciences Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; Professor, Department of Parasitology & Animal Diseases, National Research Centre, 33 Bohouth St., Dokki, Giza 12622, Egypt
| | - Abdulkarim S Binshaya
- Department of Medical Laboratory Science, College of Applied Medical Sciences Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia.
| |
Collapse
|
40
|
He Y, E M, Liu S, Liu G, Cao Y. The cellular signaling and regulatory role of protein phosphatase in tumor diagnosis: Upstream miRNAs of PTEN. Int J Biol Macromol 2024; 280:136179. [PMID: 39357725 DOI: 10.1016/j.ijbiomac.2024.136179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/25/2024] [Accepted: 09/29/2024] [Indexed: 10/04/2024]
Abstract
Protein phosphatases have demonstrated considerable promise in the realm of early tumor diagnosis across various malignancies. These enzymes play a critical role in modulating the PI3K-Akt signaling pathway, which is integral to cellular processes such as proliferation, survival, and migration. When the activity of protein phosphatases becomes abnormal, it can disrupt these essential signaling pathways, potentially leading to the initiation and progression of tumors. Consequently, monitoring for abnormal expression and activity levels of protein phosphatases could serve as a vital biomarker for early cancer detection. By identifying these alterations, clinicians may be better equipped to diagnose tumors at an earlier stage, significantly improving patient outcomes.In summary, our study highlights the multifaceted and significant role of PTEN in various forms of cancer, including esophageal squamous cell carcinoma (ESCA). Further analysis showed that the expression levels of protein phosphatase and PTEN protein were significantly associated with the early diagnosis of tumors, especially in the early stage of tumors, and their detection sensitivity and specificity were high. Therefore, by detecting the expression of protein phosphatase and PTEN protein, the early diagnosis of tumor can be achieved, and the therapeutic effect and prognosis of patients can be improved.
Collapse
Affiliation(s)
- Yunlong He
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, Heilongjiang 150060, China
| | - Mingyan E
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, Heilongjiang 150060, China.
| | - Shuang Liu
- Department of Gynecology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, Heilongjiang 150060, China
| | - Guohui Liu
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, Heilongjiang 150060, China
| | - Yang Cao
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, Heilongjiang 150060, China
| |
Collapse
|
41
|
Wang J, Tao R, Hu H, Gao J, Liu Y, Xia J, Lan X, Di Y. miR-320b, a Future Expected New Biomarker for Type 2 Diabetes Mellitus Induces Dysglycemia by Targeting PTEN. Int J Endocrinol 2024; 2024:5540062. [PMID: 39502509 PMCID: PMC11535181 DOI: 10.1155/2024/5540062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 10/06/2024] [Accepted: 10/15/2024] [Indexed: 11/08/2024] Open
Abstract
Background: Type 2 diabetes mellitus (T2DM) has emerged as a global epidemic issue, with high rates of disability and fatality. Traditional diagnostic biomarkers are typically detected once a metabolic imbalance has already occurred, thus the development of early diagnostic biomarkers is crucial for T2DM. Metabolomics studies have identified several predictive biomarkers for T2DM, including miR-320. Our previous research found that miR-320b was significantly downregulated in T2DM patients, but the underlying mechanism remains unclear. Therefore, this study was designed to investigate the significance of miR-320b for T2DM diagnosis and to explore the involved molecular mechanism. Methods: A total of 50 patients with T2DM and 80 sex- and age-matched healthy subjects were selected. The plasma miR-320b of all participations was detected by qRT-PCR and its correlations with other biomarkers of T2DM were analyzed. Besides, the expression of miR-320b in HepG2 cells was suppressed by miRNA inhibitors. Then the glucose consumption of HepG2 cells was measured. The target gene of miR-320b was predicted by four bioinformatics tools and intersected these prediction results by Venny method. The T2DM relevant target genes were identified by the GeneCards database. To ensure disease relevance, these T2DM relevant target genes were subsequently intersected with the target genes of miR-320b. Protein-protein analysis (PPI) was used to screening the gene with the most connections in these target genes. Finally, the target gene of miR-320b specific to T2DM was confirmed directly by luciferase reporter assay. The expression of target gene in HepG2 cell culture supernatant and plasma of all participations was detected. Results: Our results showed that the expression level of miR-320b was significantly lower in T2DM patients compared to the healthy controls. It was negatively correlated with fasting plasma glucose (FPG), glycated hemoglobin (HbA1C), and homeostasis model assessment of insulin resistance (HOMA-IR), but positively with HOMA-β. The glucose consumption of HepG2 cells in the miR-320b inhibitor group was significantly lower compared to inhibitor-NC and blank control group. We predicted and confirmed that phosphatase and tensin homolog (PTEN) was the direct target gene of miR-320b using Bioinformation tools and luciferase reporter assay. Moreover, the concentration of PTEN was significantly higher in the HepG2 cell culture supernatant and plasma of T2DM patients. Conclusions: Our research demonstrated a negative correlation between miR-320b and FPG, HbA1C, and HOMA-IR, while exhibiting a positive correlation with HOMA-β. Suppressing miR-320b expression would impair glucose consumption of HepG2 cells through PI3K pathway by targeting PTEN. These results suggest that miR-320b may be a potential biomarker for diagnosing T2DM and a promising target for therapeutic intervention.
Collapse
Affiliation(s)
- Jinxingyi Wang
- Department of Pharmacy, The Second Affiliated Hospital of Guizhou Medical University, Kaili 556000, China
| | - Ruyu Tao
- Department of Pharmacy, The Second Affiliated Hospital of Guizhou Medical University, Kaili 556000, China
| | - Hanshuai Hu
- Department of Pharmacy, The Second Affiliated Hospital of Guizhou Medical University, Kaili 556000, China
| | - Jiejie Gao
- Department of Pharmacy, The Second Affiliated Hospital of Guizhou Medical University, Kaili 556000, China
| | - Yang Liu
- Department of Pharmacy, The Second Affiliated Hospital of Guizhou Medical University, Kaili 556000, China
| | - Jie Xia
- Department of Pharmacy, The Second Affiliated Hospital of Guizhou Medical University, Kaili 556000, China
| | - Xue Lan
- Department of Pharmacy, The Second Affiliated Hospital of Guizhou Medical University, Kaili 556000, China
| | - Yanan Di
- Department of Pharmacy, The Second Affiliated Hospital of Guizhou Medical University, Kaili 556000, China
| |
Collapse
|
42
|
Eid N, Bhatnagar P, Chan LL, Garcia-Macia M. Suppression of hepatic steatosis in non-alcoholic steatohepatitis model by modified Xiaoyao San formula: Evidence, mechanisms and perspective. World J Hepatol 2024; 16:1388-1392. [DOI: 10.4254/wjh.v16.i10.1388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/24/2024] [Accepted: 10/11/2024] [Indexed: 11/22/2024] Open
Abstract
In this letter, we comment on a recent publication by Mei et al, in the World Journal of Hepatology, investigating the hepatoprotective effects of the modified Xiaoyao San (MXS) formula in a male rat model of non-alcoholic steatohepatitis (NASH). The authors found that MXS treatment mitigated hepatic steatosis and inflammation in the NASH model, as evidenced by the reduction in lipid droplets (LDs), fibrosis markers and lipogenic factors. Interestingly, these hepatoprotective effects were associated with androgen upregulation (based on metabolomics analysis of male steroid hormone metabolites), adenosine 5’-monophosphate-activated protein kinase (AMPK) activation, and restoration of phosphatase and tensin homolog (PTEN) expression. However, the authors did not clearly discuss the relationships between MXS-induced hepatic steatosis reduction in the NASH model, and androgen upregulation, AMPK activation, and restoration of PTEN expression. This editorial emphasizes the reported mechanisms and explains how they act or interact with each other to reduce hepatic steatosis and inflammation in the NASH model. As a perspective, we propose additional mechanisms (such as autophagy/lipophagy activation in hepatocytes) for the clearance of LDs and suppression of hepatic steatosis by MXS in the NASH model. A proper understanding of the mechanisms of MXS-induced reduction of hepatic steatosis might help in the treatment of NASH and related diseases.
Collapse
Affiliation(s)
- Nabil Eid
- Department of Anatomy, Division of Human Biology, School of Medicine, IMU University, Kuala Lumpur 57000, Malaysia
| | - Payal Bhatnagar
- Department of Pharmaceutical Technology, School of pharmacy, IMU University, Kuala Lumpur 57000, Malaysia
| | - Li-Li Chan
- Department of Pathology and Pharmacology, School of Medicine, IMU University, Kuala Lumpur 57000, Malaysia
| | - Marina Garcia-Macia
- Institute of Functional Biology and Genomics, Department of Biochemistry and Molecular Biology, University of Salamanca, Salamanca 37007, Spain
| |
Collapse
|
43
|
Eid N, Bhatnagar P, Chan LL, Garcia-Macia M. Suppression of hepatic steatosis in non-alcoholic steatohepatitis model by modified Xiaoyao San formula: Evidence, mechanisms and perspective. World J Hepatol 2024; 16:1208-1212. [PMID: 39474573 PMCID: PMC11514612 DOI: 10.4254/wjh.v16.i10.1208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/24/2024] [Accepted: 10/11/2024] [Indexed: 10/21/2024] Open
Abstract
In this letter, we comment on a recent publication by Mei et al, in the World Journal of Hepatology, investigating the hepatoprotective effects of the modified Xiaoyao San (MXS) formula in a male rat model of non-alcoholic steatohepatitis (NASH). The authors found that MXS treatment mitigated hepatic steatosis and inflammation in the NASH model, as evidenced by the reduction in lipid droplets (LDs), fibrosis markers and lipogenic factors. Interestingly, these hepatoprotective effects were associated with androgen upregulation (based on metabolomics analysis of male steroid hormone metabolites), adenosine 5'-monophosphate-activated protein kinase (AMPK) activation, and restoration of phosphatase and tensin homolog (PTEN) expression. However, the authors did not clearly discuss the relationships between MXS-induced hepatic steatosis reduction in the NASH model, and androgen upregulation, AMPK activation, and restoration of PTEN expression. This editorial emphasizes the reported mechanisms and explains how they act or interact with each other to reduce hepatic steatosis and inflammation in the NASH model. As a perspective, we propose additional mechanisms (such as autophagy/lipophagy activation in hepatocytes) for the clearance of LDs and suppression of hepatic steatosis by MXS in the NASH model. A proper understanding of the mechanisms of MXS-induced reduction of hepatic steatosis might help in the treatment of NASH and related diseases.
Collapse
Affiliation(s)
- Nabil Eid
- Department of Anatomy, Division of Human Biology, School of Medicine, IMU University, Kuala Lumpur 57000, Malaysia.
| | - Payal Bhatnagar
- Department of Pharmaceutical Technology, School of pharmacy, IMU University, Kuala Lumpur 57000, Malaysia
| | - Li-Li Chan
- Department of Pathology and Pharmacology, School of Medicine, IMU University, Kuala Lumpur 57000, Malaysia
| | - Marina Garcia-Macia
- Institute of Functional Biology and Genomics, Department of Biochemistry and Molecular Biology, University of Salamanca, Salamanca 37007, Spain
| |
Collapse
|
44
|
Wang TT, Ji YM, Zhang Q, Liang B, Fan TT, Ye X. METTL14 Induced N 6-Methyladenosine Modification of FOXP4 mRNA in HBV-HCC. J Cancer 2024; 15:6232-6238. [PMID: 39513116 PMCID: PMC11540497 DOI: 10.7150/jca.101385] [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: 07/24/2024] [Accepted: 09/28/2024] [Indexed: 11/15/2024] Open
Abstract
Chronic hepatitis B virus infections are a significant cause of liver cirrhosis and cancer. Our research reveals that HBV infection leads to a marked increase in m6A modification of Foxp4 mRNA, resulting in enhanced stability of the mRNA and a subsequent increase in Foxp4 mRNA levels. Analysis of biopsy samples from chronic HBV patients demonstrated consistent upregulation of m6A-modified Foxp4 mRNA levels alongside increased Foxp4 mRNA levels. Functionally, Foxp4 was found to promote proliferation, migration, and invasion of hepatocellular carcinoma (HCC) cells in laboratory settings. Additionally, HBV gene expression was shown to activate the PI3K/AKT pathway by modulating Foxp4 mRNA stability in HCC cells. This study provides valuable insights into the underlying mechanisms of HBV infection and its potential implications for cancer development.
Collapse
Affiliation(s)
- Tian-Tian Wang
- Department of Endoscopy, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200438, China
| | - Yi-Mei Ji
- Department of Endoscopy, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200438, China
| | - Qian Zhang
- Department of Endoscopy, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200438, China
| | - Bo Liang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang University School of Medicine, Nanchang, Jiangxi Province, China
| | - Ting-ting Fan
- Department of Endoscopy, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200438, China
| | - Xin Ye
- Department of Endoscopy, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200438, China
| |
Collapse
|
45
|
Fang H, Wang X, Wang Z, Ma X, Zhang L, Yang L. Modulation of PI3K/AKT/mTOR signaling pathway in the ovine liver and duodenum during early pregnancy. Domest Anim Endocrinol 2024; 89:106870. [PMID: 38954983 DOI: 10.1016/j.domaniend.2024.106870] [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: 03/16/2024] [Revised: 06/22/2024] [Accepted: 06/26/2024] [Indexed: 07/04/2024]
Abstract
The liver and intestine play a critical role in nutrient absorption, storage, and metabolism. The aim of this study was to evaluate expression pattern of phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of the rapamycin (mTOR) signaling pathway that included PI3K, AKT1, mTOR, FoxO1, SREBP-1, PPARα, PTEN and FXR in the maternal liver and duodenum. Ovine livers and duodenums were sampled at day 16 of the estrous cycle, and at days 13, 16 and 25 of gestation, and RT-qPCR, western blot and immunohistochemistry analysis were used to detect mRNA and protein expression. The results showed that expression of PI3K, AKT1, p-mTOR, FoxO1, SREBP-1 and PTEN upregulated in the maternal liver, and PPARα upregulated in the duodenum. However, expression of FoxO1, SREBP-1 and PTEN in the duodenum downregulated during early pregnancy. In addition, expression levels of SREBP-1, PTEN and PPARα in the maternal liver, and PI3K in the duodenum peaked at day 13 of pregnancy. In addition, expression levels of PI3K, p-mTOR and FoxO1 in the liver, and AKT1 and p-mTOR in the duodenum peaked at day 16 of pregnancy. Nevertheless, expression levels of FXR both in the maternal liver duodenum downregulated at days 13 and 16 of pregnancy. In conclusion, early pregnancy regulated expression pattern of PI3K/AKT/mTOR signaling pathway in the ovine liver and duodenum in a pregnancy stage-specific and tissue-specific manner, which may be necessary for the adaptations in maternal hepatic nutrient metabolism and intestinal nutrient absorption early pregnancy.
Collapse
Affiliation(s)
- Hongxu Fang
- School of Life Sciences and Food Engineering, Hebei University of Engineering, No. 19 Taiji Road, Handan 056038, PR China
| | - Xinxin Wang
- School of Life Sciences and Food Engineering, Hebei University of Engineering, No. 19 Taiji Road, Handan 056038, PR China
| | - Zhongyue Wang
- School of Life Sciences and Food Engineering, Hebei University of Engineering, No. 19 Taiji Road, Handan 056038, PR China
| | - Xiaoxin Ma
- School of Life Sciences and Food Engineering, Hebei University of Engineering, No. 19 Taiji Road, Handan 056038, PR China
| | - Leying Zhang
- School of Life Sciences and Food Engineering, Hebei University of Engineering, No. 19 Taiji Road, Handan 056038, PR China
| | - Ling Yang
- School of Life Sciences and Food Engineering, Hebei University of Engineering, No. 19 Taiji Road, Handan 056038, PR China.
| |
Collapse
|
46
|
You Y, Chen S, Tang B, Xing X, Deng H, Wu Y. Exosome-related gene identification and diagnostic model construction in hepatic ischemia-reperfusion injury. Sci Rep 2024; 14:22450. [PMID: 39341981 PMCID: PMC11439056 DOI: 10.1038/s41598-024-73441-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024] Open
Abstract
Hepatic ischemia-reperfusion injury (HIRI) may cause severe hepatic impairment, acute hepatic insufficiency, and multiorgan system collapse. Exosomes can alleviate HIRI. Therefore, this study explored the role of exosomal-related genes (ERGs) in HIRI using bioinformatics to determine the underlying molecular mechanisms and novel diagnostic markers for HIRI. We merged the GSE12720, GSE14951, and GSE15480 datasets obtained from the Gene Expression Omnibus (GEO) database into a combined gene dataset (CGD). CGD was used to identify differentially expressed genes (DEGs) based on a comparison of the HIRI and healthy control cohorts. The impact of these DEGs on HIRI was assessed through gene set enrichment analysis (GSEA) and gene set variation analysis (GSVA). ERGs were retrieved from the GeneCards database and prior studies, and overlapped with the identified DEGs to yield the set of exosome-related differentially expressed genes (ERDEGs). Functional annotations and enrichment pathways of these genes were determined using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses. Diagnostic models for HIRI were developed using least absolute shrinkage and selection operator (LASSO) regression and support vector machine (SVM) algorithms. Key genes with diagnostic value were identified from the overlap, and single-sample gene-set enrichment analysis (ssGSEA) was conducted to evaluate the immune infiltration characteristics. A molecular regulatory interaction network was established using Cytoscape software to elucidate the intricate regulatory mechanisms of key genes in HIRI. Finally, exosome score (Es) was obtained using ssGSEA and the HIRI group was divided into the Es_High and Es_Low groups based on the median Es. Gene expression was analyzed to understand the impact of all genes in the CGD on HIRI. Finally, the relative expression levels of the five key genes in the hypoxia-reoxygenation (H/R) model were determined using quantitative real-time PCR (qRT-PCR). A total of 3810 DEGs were identified through differential expression analysis of the CGD, and 61 of these ERDEGs were screened. Based on GO and KEGG enrichment analyses, the ERDEGs were mainly enriched in wound healing, MAPK, protein kinase B signaling, and other pathways. GSEA and GSVA revealed that these genes were mainly enriched in the TP53, MAPK, TGF[Formula: see text], JAK-STAT, MAPK, and NFKB pathways. Five key genes (ANXA1, HNRNPA2B1, ICAM1, PTEN, and THBS1) with diagnostic value were screened using the LASSO regression and SVM algorithms and their molecular interaction network was established using Cytoscape software. Based on ssGSEA, substantial variations were found in the expression of 18 immune cell types among the groups (p < 0.05). Finally, the Es of each HIRI patient was calculated. ERDEGs in the Es_High and Es_Low groups were enriched in the IL18, TP53, MAPK, TGF[Formula: see text], and JAK-STAT pathways. The differential expression of these five key genes in the H/R model was verified using qRT-PCR. Herein, five key genes were identified as potential diagnostic markers. Moreover, the potential impact of these genes on pathways and the regulatory mechanisms of their interaction network in HIRI were revealed. Altogether, our findings may serve as a theoretical foundation for enhancing clinical diagnosis and elucidating underlying pathogeneses.
Collapse
Affiliation(s)
- Yujuan You
- Department of Anesthesiology, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330008, P. R. China
| | - Shoulin Chen
- Department of Anesthesiology, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330008, P. R. China
| | - Binquan Tang
- Department of Anesthesiology, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330008, P. R. China
| | - Xianliang Xing
- Department of Anesthesiology, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330008, P. R. China
| | - Huanling Deng
- Department of Anesthesiology, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330008, P. R. China
| | - Yiguo Wu
- Department of Blood Transfusion, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330008, P. R. China.
| |
Collapse
|
47
|
Eisenreich W, Leberfing J, Rudel T, Heesemann J, Goebel W. Interactions of SARS-CoV-2 with Human Target Cells-A Metabolic View. Int J Mol Sci 2024; 25:9977. [PMID: 39337465 PMCID: PMC11432161 DOI: 10.3390/ijms25189977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/13/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Viruses are obligate intracellular parasites, and they exploit the cellular pathways and resources of their respective host cells to survive and successfully multiply. The strategies of viruses concerning how to take advantage of the metabolic capabilities of host cells for their own replication can vary considerably. The most common metabolic alterations triggered by viruses affect the central carbon metabolism of infected host cells, in particular glycolysis, the pentose phosphate pathway, and the tricarboxylic acid cycle. The upregulation of these processes is aimed to increase the supply of nucleotides, amino acids, and lipids since these metabolic products are crucial for efficient viral proliferation. In detail, however, this manipulation may affect multiple sites and regulatory mechanisms of host-cell metabolism, depending not only on the specific viruses but also on the type of infected host cells. In this review, we report metabolic situations and reprogramming in different human host cells, tissues, and organs that are favorable for acute and persistent SARS-CoV-2 infection. This knowledge may be fundamental for the development of host-directed therapies.
Collapse
Affiliation(s)
- Wolfgang Eisenreich
- Structural Membrane Biochemistry, Bavarian NMR Center (BNMRZ), Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, Lichtenbergstr. 4, 85747 Garching, Germany;
| | - Julian Leberfing
- Structural Membrane Biochemistry, Bavarian NMR Center (BNMRZ), Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, Lichtenbergstr. 4, 85747 Garching, Germany;
| | - Thomas Rudel
- Chair of Microbiology, Biocenter, University of Würzburg, 97074 Würzburg, Germany;
| | - Jürgen Heesemann
- Max von Pettenkofer Institute, Ludwig Maximilian University of Munich, 80336 München, Germany; (J.H.); (W.G.)
| | - Werner Goebel
- Max von Pettenkofer Institute, Ludwig Maximilian University of Munich, 80336 München, Germany; (J.H.); (W.G.)
| |
Collapse
|
48
|
Tian Y, Liu YF, Wang YY, Li YZ, Ding WY, Zhang C. Molecular mechanisms of PTEN in atherosclerosis: A comprehensive review. Eur J Pharmacol 2024; 979:176857. [PMID: 39094923 DOI: 10.1016/j.ejphar.2024.176857] [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: 03/09/2024] [Revised: 07/17/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024]
Abstract
Atherosclerosis is a chronic inflammatory disease of the arterial wall caused by an imbalance of lipid metabolism and a maladaptive inflammatory response. A variety of harmful cellular changes associated with atherosclerosis include endothelial dysfunction, the migration of circulating inflammatory cells to the arterial wall, the production of proinflammatory cytokines, lipid buildup in the intima, local inflammatory responses in blood vessels, atherosclerosis-associated apoptosis, and autophagy. PTEN inhibits the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (PKB/AKT)/mammalian target of rapamycin (mTOR) pathway through its lipid phosphatase activity. Previous studies have shown that PTEN is closely related to atherosclerosis. This article reviews the role of PTEN in atherosclerosis from the perspectives of autophagy, apoptosis, inflammation, proliferation, and angiogenesis.
Collapse
Affiliation(s)
- Yuan Tian
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan province, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People's Republic of China
| | - Yi-Fan Liu
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People's Republic of China
| | - Yan-Yue Wang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan province, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People's Republic of China
| | - Yong-Zhen Li
- Department of Pathology, The First People's Hospital of Zigong, Zigong, China, 643099, People's Republic of China
| | - Wen-Yan Ding
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan province, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People's Republic of China
| | - Chi Zhang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan province, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People's Republic of China.
| |
Collapse
|
49
|
James AJ, Torres-Guzman RA, Chaker SC, Sigel ME, Perdikis G, Supp DM, Dale Slater EL. Global insights into keloid formation: An international systematic review of regional genetic risk factors and commonalities. Wound Repair Regen 2024; 32:746-757. [PMID: 38958095 DOI: 10.1111/wrr.13203] [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: 02/02/2024] [Revised: 05/28/2024] [Accepted: 06/14/2024] [Indexed: 07/04/2024]
Abstract
Keloid disorder is a morbid and disfiguring benign fibroproliferative disease with a higher incidence in groups with darker skin pigmentation. Predicting keloidogenesis in patients is difficult with treatment primarily aimed at preventing further scar expansion and improving aesthetics without addressing their unknown underlying pathophysiology. We aimed to identify potential genetic predispositions to keloid scarring in the literature. A search was conducted on 21 August 2023, by the first and second authors independently from 1985 to August 2023 using PubMed, MEDLINE, Embase, Web of Science, Scopus and CINAHL. The following MeSH terms were used: 'Keloid', 'Risk' and 'Genetic'. Two researchers independently searched for studies based on titles and abstracts and screened filtered articles by reviewing full text. If no agreement could be reached, a third senior author designated whether the article should be included. We used the Preferred Reporting Items for Systematic Reviews and Meta-Analyses 2020 statement as the basis of our organisation. Human studies with genetic analysis to determine an association of a protein or gene to keloidogenesis were selected for inclusion. Studies in languages other than English, reviews, conference articles, and book chapters were excluded. Fifty studies met inclusion criteria. The human leukocyte antigen (HLA) system was broadly implicated, and the DRB1*15 allele was associated with an increased risk of keloid in three separate ethnic groups. Some HLA Class I alleles were associated with keloid in one population but not in others. Additionally, polymorphisms in the E3 ubiquitin-protein ligase (NEDD4) signal cascade and vitamin D receptor (VDR) have been implicated in diverse groups. No current genetic test can predict keloid risk. Our review identified candidate predisposing genes, including NEDD4, VDR and components of the HLA system. Further studies in heterogeneous populations are needed to identify reliable screening targets.
Collapse
Affiliation(s)
- Andrew J James
- Department of Plastic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Ricardo A Torres-Guzman
- Department of Plastic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Sara C Chaker
- Department of Plastic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | - Galen Perdikis
- Department of Plastic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Dorothy M Supp
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Shriners Children's Ohio, Dayton, Ohio, USA
| | - Elizabeth L Dale Slater
- Department of Plastic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
50
|
Fieni C, Sorrentino C, Ciummo SL, Fontana A, Lotti LV, Scialis S, Calvo Garcia D, Caulo M, Di Carlo E. Immunoliposome-based targeted delivery of the CRISPR/Cas9gRNA-IL30 complex inhibits prostate cancer and prolongs survival. Exp Mol Med 2024; 56:2033-2051. [PMID: 39232121 PMCID: PMC11447253 DOI: 10.1038/s12276-024-01310-2] [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: 02/05/2024] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 09/06/2024] Open
Abstract
The development of selective and nontoxic immunotherapy targeting prostate cancer (PC) is challenging. Interleukin (IL)30 plays immunoinhibitory and oncogenic roles in PC, and its tumor-specific suppression may have significant clinical implications. CRISPR/Cas9-mediated IL30 gene deletion in PC xenografts using anti-PSCA antibody-driven lipid nanocomplexes (Cas9gRNA-hIL30-PSCA NxPs) revealed significant genome editing efficiency and circulation stability without off-target effects or organ toxicity. Biweekly intravenous administration of Cas9gRNA-hIL30-PSCA NxPs to PC-bearing mice inhibited tumor growth and metastasis and improved survival. Mechanistically, Cas9gRNA-hIL30-PSCA NxPs suppressed ANGPTL 1/2/4, IL1β, CCL2, CXCL1/6, SERPINE1-F1, EFNB2, PLG, PF4, VEGFA, VEGFD, ANG, TGFβ1, EGF and HGF expression in human PC cells while upregulated CDH1, DKK3 and PTEN expression, leading to low proliferation and extensive ischemic necrosis. In the syngeneic PC model, IL30-targeting immunoliposomes downregulated NFKB1 expression and prevented intratumoral influx of CD11b+Gr-1+MDCs, Foxp3+Tregs, and NKp46+RORγt+ILC3, and prolonged host survival by inhibiting tumor progression. This study serves as a proof of principle that immunoliposome-based targeted delivery of Cas9gRNA-IL30 represent a potentially safe and effective strategy for PC treatment.
Collapse
Affiliation(s)
- Cristiano Fieni
- Department of Medicine and Sciences of Aging, "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
- Anatomic Pathology and Immuno-Oncology Unit, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
| | - Carlo Sorrentino
- Department of Medicine and Sciences of Aging, "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
- Anatomic Pathology and Immuno-Oncology Unit, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
| | - Stefania Livia Ciummo
- Department of Medicine and Sciences of Aging, "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
- Anatomic Pathology and Immuno-Oncology Unit, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
| | - Antonella Fontana
- Department of Pharmacy, "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
- UDA-TECHLAB Research Center, "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
| | | | - Sofia Scialis
- Anatomic Pathology and Immuno-Oncology Unit, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
| | - Darien Calvo Garcia
- Department of Neuroscience, Imaging and Clinical Sciences, "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
- Institute for Advanced Biomedical Technologies (ITAB), "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
| | - Massimo Caulo
- Department of Neuroscience, Imaging and Clinical Sciences, "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
- Institute for Advanced Biomedical Technologies (ITAB), "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
| | - Emma Di Carlo
- Department of Medicine and Sciences of Aging, "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy.
- Anatomic Pathology and Immuno-Oncology Unit, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy.
| |
Collapse
|