1
|
Ma Y, Lai P, Sha Z, Li B, Wu J, Zhou X, He C, Ma X. TME-responsive nanocomposite hydrogel with targeted capacity for enhanced synergistic chemoimmunotherapy of MYC-amplified osteosarcoma. Bioact Mater 2025; 47:83-99. [PMID: 39897587 PMCID: PMC11783017 DOI: 10.1016/j.bioactmat.2025.01.006] [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: 08/24/2024] [Revised: 01/05/2025] [Accepted: 01/07/2025] [Indexed: 02/04/2025] Open
Abstract
The oncogene MYC is one of the most commonly activated oncogenic proteins in human tumors, with nearly one-fourth of osteosarcoma showing MYC amplification and exhibiting the worst clinical outcomes. The clinical efficacy of single radiotherapy, chemotherapy, and immunotherapy for such osteosarcoma is poor, and the dysregulation of MYC amplification and immune-suppressive tumor microenvironment (TME) may be potential causes of anti-tumor failure. To address the above issues, we developed an injectable TME-responsive nanocomposite hydrogel to simultaneously deliver an effective MYC inhibitor (NHWD-870) and IL11Rα-targeted liposomes containing cisplatin-loaded MnO2 (Cis/Mn@Lipo-IL11). After in situ administration, NHWD-870 effectively degrades MYC and downregulates CCL2 and IL13 cytokines to trigger M1 type activation of macrophages. Meanwhile, targeted delivery of Cis/Mn@Lipo-IL11 reacts with excess intratumoral GSH to generate Mn2+ and thus inducing excess active oxygen species (ROS) production through Fenton-like reaction, along with cisplatin, thereby inducing immunogenic cell death (ICD) to promote dendritic cell maturation. Through synergistic regulation of MYC and ICD levels, the immune microenvironment was reshaped to enhance immune infiltration. In the osteosarcoma-bearing model, the nanocomposite hydrogel significantly enhanced tumor T cell infiltration, induced effective anti-tumor immunity and attenuated lung metastasis. Therefore, our results reveal a powerful strategy for targeted combination therapy of MYC-amplified osteosarcoma.
Collapse
Affiliation(s)
- Yichao Ma
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Peng Lai
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Zhou Sha
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, 200092, China
| | - Bing Li
- College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Jiangpeng Wu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Xiaojun Zhou
- College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Chuanglong He
- College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Xiaojun Ma
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| |
Collapse
|
2
|
Shao F, Wang R, Li X, Hu Y, Zhang Z, Cai J, Yang J, Feng X, Ren S, Huang Z, Xie Y. TTC36 promotes proliferation and drug resistance in hepatocellular carcinoma cells by inhibiting c-Myc degradation. Cell Death Dis 2025; 16:332. [PMID: 40274799 DOI: 10.1038/s41419-025-07663-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 04/08/2025] [Accepted: 04/11/2025] [Indexed: 04/26/2025]
Abstract
High c-Myc protein accumulation contributes to the proliferation, invasion, and drug resistance in multiple cancer cells, but the underlying mechanism about c-Myc accumulation remains not to be elucidated. Here, we demonstrate that TTC36 promotes c-Myc protein accumulation in hepatocellular carcinoma cells, thereby driving the proliferation and sorafenib resistance in hepatocellular carcinoma cells. Ttc36 depletion disrupts the interaction between SET and PPP2R1A, consequently activating PP2A. Activated PP2A directly dephosphorylates p-c-MycS62 and activates GSK3β, relying on AKT, leading increased phosphorylation of p-c-MycT58, finally promotes FBXW7-mediated polyubiquitination and degradation of c-Myc. Inhibitors targeting GSK3β and PP2A effectively reverse the sorafenib resistance promoted by TTC36. These findings highlight the crucial role of TTC36 in c-Myc accumulation-caused proliferation and sorafenib resistance in HCC, providing a promising combination strategy for treating patients with c-Myc protein accumulation in advanced HCC.
Collapse
Affiliation(s)
- Fengling Shao
- The Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Runzhi Wang
- The Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Xinyi Li
- The Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Yanxia Hu
- School of Life and Health Sciences, Hainan University, Haikou, China
| | - Zaikuan Zhang
- The Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Jing Cai
- College of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Jieru Yang
- The Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Xiaosong Feng
- The Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Suxia Ren
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China.
| | - Zengyi Huang
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China.
- Mitomedical laboratory of Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China.
| | - Yajun Xie
- The Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
3
|
Bai S, Hu Y, Chen N, Zhou L, Ju W, Qiao X, Yu J. DCAF13 influences breast cancer chemotherapy resistance through metabolic reprogramming by regulating c-Myc expression. Med Oncol 2025; 42:178. [PMID: 40268788 DOI: 10.1007/s12032-025-02722-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 04/14/2025] [Indexed: 04/25/2025]
Abstract
Chemotherapy resistance remains a major obstacle in breast cancer treatment. This study identifies DCAF13, a substrate recognition receptor of the CRL4 ubiquitin ligase complex, as a critical regulator of chemotherapy resistance through c-Myc-driven metabolic reprogramming. We found that DCAF13 expression was significantly upregulated in chemotherapy-resistant breast cancer cell lines compared to their parental counterparts. Inhibition of DCAF13 enhanced chemotherapy sensitivity, whereas its overexpression suppressed drug sensitivity. Mechanistically, DCAF13 upregulated c-Myc expression, driving metabolic reprogramming, characterized by increased glycolysis and oxidative phosphorylation. This metabolic shift promoted cell proliferation and resistance to chemotherapy. Clinically, DCAF13 expression correlated with poor prognosis in breast cancer patients, particularly in advanced stages and triple-negative breast cancer (TNBC). Our findings highlight the DCAF13-c-Myc axis as a critical mediator of chemotherapy resistance, suggesting that targeting this pathway could provide novel therapeutic strategies to overcome drug resistance in breast cancer. Further clinical studies are needed to explore the potential of DCAF13 as a therapeutic target.
Collapse
Affiliation(s)
- Shiqiang Bai
- Department of Pathology, The 966th Hospital of The PLA Joint Logistic Support Force, Dandong, 118000, China
| | - Yunlong Hu
- Department of Emergency and Intensive Care Unit, The 966th Hospital of The PLA Joint Logistic Support Force, Dandong, 118000, China
| | - Ning Chen
- Department of Pathology, The 966th Hospital of The PLA Joint Logistic Support Force, Dandong, 118000, China
| | - Liang Zhou
- Department of General Surgery, The 966th Hospital of The PLA Joint Logistic Support Force, Dandong, 118000, China
| | - Weiwei Ju
- Medical College of Liaodong University, Dandong, 118003, China
| | - Xinyi Qiao
- Department of General Surgery, The 966th Hospital of The PLA Joint Logistic Support Force, Dandong, 118000, China
| | - Jianyu Yu
- Department of General Surgery, The 966th Hospital of The PLA Joint Logistic Support Force, Dandong, 118000, China.
| |
Collapse
|
4
|
Huang C, Harris KS, Siddiqui G, Jörg M. Recommended Tool Compounds: Thienotriazolodiazepines-Derivatized Chemical Probes to Target BET Bromodomains. ACS Pharmacol Transl Sci 2025; 8:978-1012. [PMID: 40242580 PMCID: PMC11997894 DOI: 10.1021/acsptsci.4c00726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 01/29/2025] [Accepted: 02/18/2025] [Indexed: 04/18/2025]
Abstract
Thienotriazolodiazepines, including (+)-JQ1 (4), are well-known inhibitors of the bromodomain (BD) and extra-terminal domain (BET) family of proteins. Despite the suboptimal physicochemical properties as a drug candidate, such as poor solubility and half-life, (+)-JQ1 (4) has proven as an effective chemical probe with high target potency and selectivity. (+)-JQ1 (4) and (+)-JQ1-derived chemical probes have played a vital role in chemical biology and drug discovery over the past decade, which is demonstrated by the high number of impactful research studies published since the disclosure of (+)-JQ1 (4) in 2010. In this review, we discuss the development of (+)-JQ1-derivatized chemical probes over the past decade and their significant contribution to scientific research. Specifically, we will summarize the development of innovative label-free and labeled (+)-JQ1-derivatized chemical probes, such as bivalent, covalent, and photoaffinity probes as well as protein degraders, with a focus on the design of these chemical probes.
Collapse
Affiliation(s)
- Chuhui Huang
- Medicinal
Chemistry, Monash Institute of Pharmaceutical Science, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
- Drug
Delivery, Disposition & Dynamics, Monash Institute of Pharmaceutical
Science, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Kate S. Harris
- Chemistry-School
of Natural and Environmental Sciences, Newcastle
University, Newcastle
Upon Tyne NE1 7RU, United Kingdom
| | - Ghizal Siddiqui
- Drug
Delivery, Disposition & Dynamics, Monash Institute of Pharmaceutical
Science, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Manuela Jörg
- Medicinal
Chemistry, Monash Institute of Pharmaceutical Science, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
- Chemistry-School
of Natural and Environmental Sciences, Newcastle
University, Newcastle
Upon Tyne NE1 7RU, United Kingdom
| |
Collapse
|
5
|
Li L, Xie S, Zhou J, Ran J. Utilizing aptamers in targeted protein degradation strategies for disease therapy. J Pathol 2025. [PMID: 40207978 DOI: 10.1002/path.6422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/27/2025] [Accepted: 02/26/2025] [Indexed: 04/11/2025]
Abstract
Targeted protein degradation (TPD) has emerged as a promising therapeutic strategy, offering the potential to reduce disease-causing proteins that have traditionally been challenging to target using conventional small molecules. Despite significant advances made with TPD technologies, challenges such as high molecular weight, difficulties in identifying suitable ligands, suboptimal absorption, and metabolic instability remain unresolved. Recently, aptamers - single-stranded DNA or RNA oligonucleotides known for their high specificity and affinity for protein targets - have introduced novel opportunities to expand the scope of TPD, a strategy now referred to as aptamer-based TPD. This approach has demonstrated considerable promise in treating various diseases, such as cancer and ocular disorders. For example, an aptamer-proteolysis-targeting chimera (PROTAC) conjugate (APC) improved tumor targeting and reduced toxicity in a breast cancer model, and a vascular endothelial growth factor-degrading (VED)-lysosome-targeting chimera (LYTAC) molecule effectively inhibited abnormal vascular growth in vascular retinal diseases. These examples highlight the practical relevance and potential in advancing drug discovery efforts. In this review we provide a comprehensive overview of the latest advances in aptamer-based TPD strategies, including proteolysis-targeting and lysosome-targeting chimeras, emphasizing their applications, potential therapeutic benefits, as well as the challenges that must be overcome to fully harness their clinical potential. © 2025 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Lin Li
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, PR China
| | - Songbo Xie
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, PR China
| | - Jun Zhou
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, PR China
- Department of Genetics and Cell Biology, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, Nankai University, Tianjin, PR China
| | - Jie Ran
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, PR China
| |
Collapse
|
6
|
Liu T, Cui Y, Ouyang Y, Wang M, Yue S. Exosomal CCT3 as a biomarker for diagnosis and immune therapy response in patients diagnosed with hepatocellular carcinoma. Dig Liver Dis 2025:S1590-8658(25)00301-9. [PMID: 40221386 DOI: 10.1016/j.dld.2025.03.022] [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: 07/24/2024] [Revised: 11/19/2024] [Accepted: 03/21/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the dominant type of liver cancer and is associated with a high mortality rate. However, HCC lacks biomarkers for diagnosis and immune therapy response. Tumor-derived exosomes (TDEs) carcinogen-specific molecules have been used for screening multiple biomarkers. This study aimed to identify new biomarkers for the diagnosis of HCC and response to immune checkpoint blockade (ICB) therapy. METHODS Analysis of differentially expressed genes (DEGs) in HCC and normal tissues was integrated using The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO), and ExoCarta datasets. The expression of CCT3 was validated in samples from patients with HCC using quantitative polymerase chain reaction (qPCR), Western blotting, and immunohistochemistry (IHC) techniques. RESULTS Exosomal CCT3 was identified as a potential biomarker with significant impact. The expression of CCT3 in different tumor stages and normal tissues adjacent to the tumors (NATs) was validated using qPCR, western blotting, and IHC. CCT3 expression significantly increased the number of activated natural killer cells in HCC, as confirmed by qPCR and IHC. CCT3 expression significantly increases the expression of immune checkpoints in HCC. HCC-derived exosomes significantly increase the enrichment of CCT3. CONCLUSION Exosomal CCT3 is a biomarker for diagnosis and ICB therapy of HCC via MYC pathway activation and immune infiltration.
Collapse
Affiliation(s)
- Tiange Liu
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China; Nankai University Affiliated Eye Hospital, Nankai University, Tianjin, China; Tianjin Eye Hospital, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin, China
| | - Yanyan Cui
- The Affiliated Hospital of Chifeng University, Chifeng, Inner Mongolia, China
| | - Yiben Ouyang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Meilin Wang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Shijing Yue
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China.
| |
Collapse
|
7
|
Lauer UM, Awada A, Postel-Vinay S, Shapiro GI, Thieblemont C, Piha-Paul SA, Paik PK, Shepard DR, Docampo LI, Galot R, Rottey S, Sadrolhefazi B, Marzin K, Musa H, Schöffski P. Final results from the phase Ia/Ib study of the novel bromodomain and extra-terminal domain inhibitor, BI 894999, in patients with advanced solid tumors or diffuse large B-cell lymphoma. ESMO Open 2025; 10:104499. [PMID: 40203516 PMCID: PMC12005229 DOI: 10.1016/j.esmoop.2025.104499] [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: 08/01/2024] [Revised: 12/19/2024] [Accepted: 02/09/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Bromodomain and extraterminal domain (BET) inhibitors have demonstrated efficacy in solid and hematological malignancies. BI 894999, a novel, orally administered BET inhibitor, has demonstrated preclinical efficacy. METHODS This was an open-label, dose-finding study evaluating BI 894999 for diffuse large B-cell lymphoma (DLBCL; phase Ia extension) and solid tumors [colorectal cancer (CRC), nuclear protein in testis (NUT) carcinoma, metastatic castration-resistant prostate cancer (mCRPC) and small-cell lung cancer (SCLC); phase Ib cohort]. The primary endpoint was dose-limiting toxicities (DLTs) during the maximum tolerated dose (MTD) period (phase Ia) and treatment period (phase Ib). RESULTS Eighteen patients with DLBCL were enrolled in the phase Ia extension and 79 with solid tumors in phase Ib cohorts (SCLC, n = 12; CRC, n = 14; mCRPC, n = 11; NUT carcinoma, n = 42). Four patients had DLTs in phase Ia and 17 in phase Ib; the most frequent was grade 4 thrombocytopenia. The MTD for DLBCL was 1.5 mg (days 1-14/21). One patient (5.6%) with DLBCL achieved a partial response (PR) and three (16.7%) had stable disease. Of 42 patients with NUT carcinoma, 3 patients (7.1%) had responses (complete response, n = 1; confirmed PR, n = 1; unconfirmed PR, n = 1). Responses in other solid tumor types (n = 37) included one patient (2.7%) with mCRPC who had a confirmed PR. CONCLUSIONS The safety profile of BI 894999 was consistent with those of other BET inhibitors. Due to minimal efficacy results, further evaluation of BI 894999 as monotherapy is not planned.
Collapse
Affiliation(s)
- U M Lauer
- Department of Internal Medicine VIII, Medical Oncology & Pneumology, University Hospital Tübingen, Tübingen, Germany; German Cancer Research Center (DKFZ), Member of the German Cancer Consortium (DKTK), Tübingen, Germany.
| | - A Awada
- Oncology Medicine Department, Jules Bordet Institute, Brussels, Belgium
| | - S Postel-Vinay
- Drug Development Department (DITEP), Gustave Roussy, Villejuif, France; INSERM Unit U981, Gustave Roussy, Villejuif, France; University College of London Cancer Institute, London, UK
| | - G I Shapiro
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, USA
| | - C Thieblemont
- Université Paris Cité & Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Louis, Hémato-oncologie, Paris, France
| | - S A Piha-Paul
- Department of Investigational Cancer Therapeutics, University of Texas, MD Anderson Cancer Center, Houston, USA
| | - P K Paik
- Thoracic Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - D R Shepard
- Department of Hematology & Medical Oncology, Cleveland Clinic, Cleveland, USA
| | - L I Docampo
- Lung and Head and Neck Cancer Unit, Medical Oncology Department, University Hospital 12 de Octubre, Madrid, Spain
| | - R Galot
- Department of Medical Oncology, Institut Roi Albert II, Cliniques Universitaires Saint-Luc, Brussels, Belgium; Institute for Experimental and Clinical Research (IREC, pôle MIRO), Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - S Rottey
- Drug Research Unit Ghent, Ghent University Hospital, Ghent, Belgium
| | - B Sadrolhefazi
- Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, USA
| | - K Marzin
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - H Musa
- Boehringer Ingelheim International GmbH, Ingelheim am Rhein, Germany
| | - P Schöffski
- Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium
| |
Collapse
|
8
|
Yamamoto A, Tanaka Y, Ishibashi S, Ikeda M, Sugita K, Ono M, Nishi H, Kurata M. Modified screening of MYC promotor region elements using the CRISPR library in ovarian cancer. J Ovarian Res 2025; 18:68. [PMID: 40176181 PMCID: PMC11963443 DOI: 10.1186/s13048-025-01644-z] [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: 06/21/2024] [Accepted: 03/10/2025] [Indexed: 04/04/2025] Open
Abstract
Ovarian cancer remains one of the most lethal gynecological malignancies owing to its high recurrence rate and chemotherapeutic resistance. MYC is a well-known proto-oncogene that is frequently amplified in ovarian cancer and has been implicated in drug resistance. Previously, we established a new promoter-reporter system combined with a CRISPR activation library to identify unknown MYC regulators, and M1AP was identified as a novel MYC regulator. However, considering the insufficient explanation for the absence of guide RNA (gRNA) of MYC, this present study explored methods to prevent the gRNA of MYC itself from binding. This study first modified the promoter-reporter vector to improve its quality, then conducted CRISPR screening and analyzed candidate genes as MYC promoter regulators using next-generation sequencing in OVSAHO ovarian cancer cells. Eighty-six genes had ≥ 1000 reads, and Pearson's correlation coefficient analysis was performed on the cBioPortal of the Cancer Genomics database. Fourteen genes were identified as candidate MYC regulators with positive and significant correlations with MYC. Seven genes, including CYP4v2, ASPH, ANP32D, PCED1A, ABI1, FUZ, and HOOK2, demonstrated significantly higher luciferase activity than the control genes. Four genes, including ABI1, PCED1A, HOOK2, and CYP4v2, activated the MYC promoter, which showed over twofold higher activity than the control when overexpressed using a vector. In conclusion, four genes that activate MYC promoters were identified in an ovarian cancer cell line using the CRISPR library system with a modified promoter-reporter tool. These results will prove helpful in the development of novel treatment strategies for ovarian cancer.
Collapse
Affiliation(s)
- Akiko Yamamoto
- Department of Obstetrics and Gynecology, Tokyo Medical University, 6-7-1Shinjuku-Ku, Tokyo , Nishishinjuku, 1600023, Japan.
- Department of Comprehensive Pathology, Graduate School of Institute of Science Tokyo, Tokyo, Japan.
| | - Yosuke Tanaka
- Bioresource Research Center, Institute of Science Tokyo, Tokyo, Japan
| | - Sachiko Ishibashi
- Department of Comprehensive Pathology, Graduate School of Institute of Science Tokyo, Tokyo, Japan
| | - Masumi Ikeda
- Department of Comprehensive Pathology, Graduate School of Institute of Science Tokyo, Tokyo, Japan
| | - Keisuke Sugita
- Department of Comprehensive Pathology, Graduate School of Institute of Science Tokyo, Tokyo, Japan
- Department of Pathology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Masanori Ono
- Department of Obstetrics and Gynecology, Tokyo Medical University, 6-7-1Shinjuku-Ku, Tokyo , Nishishinjuku, 1600023, Japan
| | - Hirotaka Nishi
- Department of Obstetrics and Gynecology, Tokyo Medical University, 6-7-1Shinjuku-Ku, Tokyo , Nishishinjuku, 1600023, Japan
| | - Morito Kurata
- Department of Comprehensive Pathology, Graduate School of Institute of Science Tokyo, Tokyo, Japan
| |
Collapse
|
9
|
Xiao Z, Wang Y, Pan D, Liu X, Gan J, Huang L, Feng Y. USP3 promotes clear cell renal cell carcinoma progression by stabilizing MYC and enhancing glycolysis. Biochim Biophys Acta Gen Subj 2025; 1869:130801. [PMID: 40164288 DOI: 10.1016/j.bbagen.2025.130801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/18/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025]
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most prevalent type of renal malignancy, and the deubiquitinase USP3 has been implicated as a critical factor in tumor biology. However, the precise mechanisms by which USP3 contributes to ccRCC progression remain unclear. This study investigates the role of USP3 in ccRCC and elucidates its underlying molecular mechanisms. Data from TCGA and GTEx databases showed elevated USP3 expression in ccRCC tissues and cell lines compared to normal renal tissues. Further analysis using qPCR and Western blot confirmed this upregulation in ccRCC cell lines. Functional assays revealed that silencing USP3 significantly impaired cell proliferation, migration, and invasion, while promoting apoptosis. Additionally, co-immunoprecipitation assays demonstrated an interaction between USP3 and MYC, with subsequent ubiquitination assays showing that USP3 regulates MYC stability. USP3 depletion also led to alterations in glycolysis-related gene expression, which could be partially reversed by MYC overexpression. These findings suggest that USP3 modulates ccRCC progression by stabilizing MYC, highlighting its potential as a therapeutic target in ccRCC treatment.
Collapse
Affiliation(s)
- Zhiliang Xiao
- Department of Urology, Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330000, China
| | - Yuan Wang
- Guangzhou Medical University, Guangzhou 511436, China
| | - Dehua Pan
- Department of Urology, Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330000, China
| | - Xin Liu
- Department of Urology, Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330000, China
| | - Jin Gan
- Department of Urology, Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330000, China
| | - Liang Huang
- Department of Urology, Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330000, China
| | - Yan Feng
- Department of Urology, Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330000, China.
| |
Collapse
|
10
|
Obisesan OA, Ofori S, Orobator ON, Sharma H, Groetecke E, Awuah SG. Discovery of a Pyrazolopyridinone-Based MYC Inhibitor That Selectively Engages Intracellular c-MYC and Disrupts MYC-MAX Heterodimerization. J Med Chem 2025; 68:6233-6251. [PMID: 40077826 DOI: 10.1021/acs.jmedchem.4c02556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
c-MYC is an oncogenic transcription factor that plays a crucial role in the regulation of downstream targets involved in proliferation, apoptosis, differentiation, metabolism, signaling, and immune response processes whose deregulation leads to the progression of different pathologies. The development of selective and potent small-molecule inhibitors of c-MYC remains a grand challenge in chemical biology and medicine due to its undruggability, derived from extensive intrinsic disorder. In this study, we identified a novel dihydro pyrazolo pyridinone scaffold, MY05, that selectively targets c-MYC in cells and disrupts MYC-MAX interaction. MY05 engages intracellular c-MYC, modulates c-MYC thermal stability, reduces c-MYC transcriptional targets, and inhibits proliferation in cancer cells and tumor growth in mice. In summary, we identified a novel compound that directly interacts with c-MYC to disrupt the transcriptional program.
Collapse
Affiliation(s)
- Oluwatosin A Obisesan
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Samuel Ofori
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Owamagbe N Orobator
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Himanshi Sharma
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Emma Groetecke
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Samuel G Awuah
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
- Center for Pharmaceutical Research and Innovation, Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536, United States
- Markey NCI Comprehensive Cancer Center, University of Kentucky, Lexington, Kentucky 40536, United States
- University of Kentucky Bioelectronics and Nanomedicine Research Center, Lexington, Kentucky 40506, United States
| |
Collapse
|
11
|
Ulug ME, Ikram S, Sayyah E, Durdağı S. Employing steered MD simulations for effective virtual screening: Active pharmacophore search by dynamic corrections to target MKK3-MYC interactions. Int J Biol Macromol 2025; 310:142602. [PMID: 40157686 DOI: 10.1016/j.ijbiomac.2025.142602] [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: 07/24/2024] [Revised: 02/05/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
The protein-protein interaction (PPI) between mitogen-activated protein kinase kinase 3 (MKK3), and MYC is a crucial regulator of oncogenic signaling, particularly in triple-negative breast cancer (TNBC). Despite its clinical significance, effective small molecule inhibitors targeting this interaction remain elusive. In this study, we employed a comprehensive in silico approach integrating dynamic structure-based pharmacophore modeling, virtual screening, molecular docking, and molecular dynamics (MD) simulations to identify potential inhibitors disrupting the MKK3-MYC interaction. The pharmacophore-based screening of over 2 million compounds from ChemDiv and Enamine libraries led to the identification of 16,766 hits, which were further refined through docking and MD-based analyses. The top-ranked molecules underwent steered molecular dynamics (sMD) simulations to evaluate the mechanical stability of their binding interactions, followed by binding free energy calculations (MM/GBSA) to assess their affinity. Notably, several hit compounds exhibited stronger binding affinities and mechanical stability compared to the reference inhibitor SGI-1027, with Z332428622, 4476-2273, and 4292-0516 emerging as the most promising candidates. The lead compounds demonstrated stable interactions with key residues at the interface of MKK3 and MYC, suggesting their potential as novel modulators of MYC-driven malignancies. These findings provide a strong computational foundation for further experimental validation and offer promising candidates for targeted therapy development in MYC-dependent cancers.
Collapse
Affiliation(s)
- Muhammet Eren Ulug
- Computational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahçeşehir University, Istanbul, Turkey; Lab for Innovative Drugs (Lab4IND), Computational Drug Design Center (HITMER), Bahçeşehir University, İstanbul, Türkiye
| | - Saima Ikram
- Computational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahçeşehir University, Istanbul, Turkey; Lab for Innovative Drugs (Lab4IND), Computational Drug Design Center (HITMER), Bahçeşehir University, İstanbul, Türkiye
| | - Ehsan Sayyah
- Computational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahçeşehir University, Istanbul, Turkey; Lab for Innovative Drugs (Lab4IND), Computational Drug Design Center (HITMER), Bahçeşehir University, İstanbul, Türkiye
| | - Serdar Durdağı
- Computational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahçeşehir University, Istanbul, Turkey; Lab for Innovative Drugs (Lab4IND), Computational Drug Design Center (HITMER), Bahçeşehir University, İstanbul, Türkiye; Molecular Therapy Lab, Department of Pharmaceutical Chemistry, School of Pharmacy, Bahçeşehir University, Istanbul, Turkey.
| |
Collapse
|
12
|
Murwanti R, Ritmaleni, Ujiantari NSO, Putra IMR, Wahyudi AF, Arifka VI. Bioinformatics study and cytotoxicity of several curcumin analogues in ovarian cancer. Curr Res Toxicol 2025; 8:100230. [PMID: 40236999 PMCID: PMC11999365 DOI: 10.1016/j.crtox.2025.100230] [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: 09/13/2024] [Revised: 03/15/2025] [Accepted: 03/17/2025] [Indexed: 04/17/2025] Open
Abstract
Ovarian cancer ranks as Indonesia's third-leading cause of cancer-related death, emphasising the need for innovative treatments. This study combined bioinformatics, molecular docking, and experimental assays to tackle this challenge. We identified 166 ovarian cancer-related genes, with MYC standing out as a key target. Analysis of MYC mutations revealed prevalent alterations, though no significant survival differences were observed in patients with or without the mutations. Molecular docking pinpointed compound B155 as a promising MYC inhibitor. A preliminary cytotoxicity assay revealed compound B155's notable activity, with an 87.19 % inhibition of cell viability at 50 μM. Most of the other curcumin analogues only caused more than 50 % inhibition at the same concentration. This result suggests alternative mechanisms of action, possibly antioxidant effects, warranting further exploration. In summary, this study unveiled MYC as a prime target for ovarian cancer treatment, with curcumin analogues like B155 showing potential. Nonetheless, the complex factors affecting cytotoxicity underscore the need for deeper investigation into these compounds' mechanisms in ovarian cancer cells.
Collapse
Affiliation(s)
- Retno Murwanti
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
- Curcumin Research Center, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | - Ritmaleni
- Curcumin Research Center, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | - Navista Sri Octa Ujiantari
- Curcumin Research Center, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | | | | | | |
Collapse
|
13
|
Wang K, Saniei S, Poddar N, Autar S, Carcamo S, Sreenath M, Peplinski JH, Ries RE, Martinez IG, Chao C, Mei AHC, Rahman N, Mekerishvili L, Quijada-Álamo M, Freed G, Zhang M, Lachman K, Diaz Z, Gonzalez MM, Zhang J, Pham G, Filipescu D, Berisa M, Balestra T, Reisz JA, D'Alessandro A, Puleston DJ, Bernstein E, Chipuk JE, Wunderlich M, Tasian SK, Marcellino BK, Glass IA, Sturgeon CM, Landau DA, Chen Z, Papapetrou EP, Izzo F, Meshinchi S, Hasson D, Wagenblast E. Ontogeny Dictates Oncogenic Potential, Lineage Hierarchy, and Therapy Response in Pediatric Leukemia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.19.643917. [PMID: 40166161 PMCID: PMC11957141 DOI: 10.1101/2025.03.19.643917] [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: 04/02/2025]
Abstract
Accumulating evidence links pediatric cancers to prenatal transformation events, yet the influence of the developmental stage on oncogenesis remains elusive. We investigated how hematopoietic stem cell developmental stages affect leukemic transformation, disease progression, and therapy response using a novel, humanized model of NUP98∷NSD1-driven pediatric acute myeloid leukemia, that is particularly aggressive with WT1 co-mutations. Fetal-derived hematopoietic stem cells readily transform into leukemia, and WT1 mutations further enhance stemness and alter lineage hierarchy. In contrast, stem cells from later developmental stages become progressively resistant to transformation. Single-cell analyses revealed that fetal-origin leukemia stem cells exhibit greater quiescence and reliance on oxidative phosphorylation than their postnatal counterparts. These differences drive distinct therapeutic responses, despite identical oncogenic mutations. In patients, onco-fetal transcriptional programs correlate with worse outcomes. By targeting key vulnerabilities of fetal-origin leukemia cells, we identified combination therapies that significantly reduce aggressiveness, highlighting the critical role of ontogeny in pediatric cancer treatment.
Collapse
Affiliation(s)
- Ke Wang
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shayan Saniei
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nikita Poddar
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute Bioinformatics for Next Generation Sequencing (BiNGS) core, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Subrina Autar
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Saul Carcamo
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute Bioinformatics for Next Generation Sequencing (BiNGS) core, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Meghana Sreenath
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jack H Peplinski
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Rhonda E Ries
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Isabella G Martinez
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Clifford Chao
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anna Huo-Chang Mei
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Noshin Rahman
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Levan Mekerishvili
- Division of Hematology and Medical Oncology, Department of Medicine and Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Physiology, Biophysics and Systems Biology Graduate Program, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Miguel Quijada-Álamo
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Grace Freed
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mimi Zhang
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Katherine Lachman
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zayna Diaz
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Manuel M Gonzalez
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jing Zhang
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Giang Pham
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dan Filipescu
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mirela Berisa
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tommaso Balestra
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Julie A Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Daniel J Puleston
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emily Bernstein
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jerry E Chipuk
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mark Wunderlich
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Sarah K Tasian
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics and Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Bridget K Marcellino
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ian A Glass
- Department of Pediatrics, University of Washington, WA, USA
| | - Christopher M Sturgeon
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dan A Landau
- Division of Hematology and Medical Oncology, Department of Medicine and Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Zhihong Chen
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eirini P Papapetrou
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Franco Izzo
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Soheil Meshinchi
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Dan Hasson
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute Bioinformatics for Next Generation Sequencing (BiNGS) core, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Elvin Wagenblast
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mindich Child Health & Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
14
|
Loganathan T, George Priya Doss C. Computational molecular insights into ibrutinib as a potent inhibitor of HER2-L755S mutant in breast cancer: gene expression studies, virtual screening, docking, and molecular dynamics analysis. Front Mol Biosci 2025; 12:1510896. [PMID: 40177517 PMCID: PMC11962039 DOI: 10.3389/fmolb.2025.1510896] [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: 10/15/2024] [Accepted: 02/24/2025] [Indexed: 04/05/2025] Open
Abstract
Background The proposed study integrates several advanced computational techniques to unravel the molecular mechanisms underlying breast cancer progression and drug resistance. Methods We investigated HER2-L755S mutation through a multi-step approach, including gene expression analysis, molecular docking, and molecular dynamics simulations. Results and Discussion By conducting a network-based analysis of gene expression data from breast cancer samples, key hub genes such as MYC, EGFR, CDKN2A, ERBB2, CDK1, E2F1, TOP2A, MDM2, TGFB1, and FOXM1 were identified, all of which are critical in tumor growth and metastasis. The study mainly focuses on the ERBB2 gene, which encodes the HER2 protein, and its common mutation HER2-L755S, associated with breast cancer and resistance to the drug lapatinib. The HER2-L755S mutation contributes to both tumorigenesis and therapeutic failure. To address this, alternative therapeutic strategies were investigated using combinatorial computational approaches. The stability and flexibility of the HER2-L755S mutation were evaluated through comparative molecular dynamics simulations over 1000 ns using Gromacs in the unbound (Apo) state. Virtual screening with Schrodinger Glide identified ibrutinib as a promising alternative to lapatinib for targeting the HER2-L755S mutant. Detailed docking and molecular dynamics simulations in the bound (Holo) state demonstrated that the HER2-L755S-ibrutinib complex exhibited higher binding affinity and lower binding energy, indicating more stable interactions compared to other complexes. MM-PBSA analysis revealed that the HER2-L755S-ibrutinib complex had more negative binding energy than the HER2-L755S-afatinib, HER2-L755S-lapatinib, and HER2-L755S-neratinib complexes, suggesting that ibrutinib forms the most stable complex with favorable binding interactions. Conclusion These results provide in-depth atomic-level insights into the binding mechanisms of these inhibitors, highlighting ibrutinib as a potentially effective inhibitor for the clinical treatment of breast cancer.
Collapse
Affiliation(s)
| | - C. George Priya Doss
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of BioSciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| |
Collapse
|
15
|
Wang L, Wang Y, Xu L. Overexpression of lncRNA TINCR inhibits cutaneous squamous cell carcinoma cells through promotes methylation of Myc and TERC genes. Arch Dermatol Res 2025; 317:559. [PMID: 40072633 PMCID: PMC11903621 DOI: 10.1007/s00403-025-03964-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/31/2024] [Accepted: 02/03/2025] [Indexed: 03/14/2025]
Abstract
Long non-coding RNA (lncRNA) TINCR has been shown to play a crucial regulatory role in various tumors. However, its specific mechanism of action in cutaneous squamous cell carcinoma (CSCC) remains unclear. This study aimed to explore the role of lncRNA TINCR in CSCC. We utilized overexpression techniques to study the effects of TINCR on CSCC cells. Methylation-specific PCR (MSP) and RNA immunoprecipitation (RIP) assays were used to assess the impact of TINCR on the methylation of the promoter regions of the Myc and TERC genes, and its interaction with DNA methyltransferase 1 (DNMT1). The results showed that overexpression of TINCR significantly promoted methylation in the promoter regions of Myc (N-MYC, L-MYC, and c-MYC) and TERC genes, inhibiting the proliferation, migration, and invasion of CSCC cells. MSP and RIP experiments further confirmed that TINCR binds to DNMT1, enhancing the methylation levels of the promoter regions of Myc and TERC genes. These findings suggest that lncRNA TINCR may serve as a potential therapeutic target for CSCC by regulating the methylation of key oncogenes. These findings provide new insights into the molecular mechanisms of CSCC and highlight the therapeutic potential of targeting TINCR.
Collapse
Affiliation(s)
- Liang Wang
- Department of Dermatology, The First Affiliated Hospital of Harbin Medical University, 23 Post Street, Nangang District, Harbin, Heilongjiang, 150001, China.
| | - Yu Wang
- Department of Dermatology, The First Affiliated Hospital of Harbin Medical University, 23 Post Street, Nangang District, Harbin, Heilongjiang, 150001, China
| | - Lei Xu
- Department of Dermatology, The First Affiliated Hospital of Harbin Medical University, 23 Post Street, Nangang District, Harbin, Heilongjiang, 150001, China
| |
Collapse
|
16
|
Ding L, Ni W, Ma Y, Xu L, Zhang Z, Liao K, Li J, Mei X, Wang Z, Ge H, Li J, Tang D, Zhang X. PDCD11 Stabilizes C‐MYC Oncoprotein by Hindering C‐MYC‐SKP2 Negative Feedback Loop to Facilitate Progression of p53‐Mutant Breast and Colon Malignancies. ADVANCED SCIENCE 2025. [DOI: 10.1002/advs.202502416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Indexed: 04/02/2025]
Abstract
AbstractC‐MYC is a proto‐oncoprotein whose dysregulation triggers tumorigenesis and tumor progression in ≈70% of cancer cases. It is presently demonstrated that aberrantly upregulated MYC is caused by the overexpressed and “extra‐nucleolar” PDCD11 in p53‐mutant breast and colon cancer cells, which is highly correlated to tumor progression, metastasis, and recurrence. In the nucleoplasm, PDCD11 binds to the TAD of C‐MYC to prevent SKP2, a transcriptional target of C‐MYC as well as one of the major E3 ligase components targeting C‐MYC, from interacting with and ubiquitinating C‐MYC in feedback. The ensuing stabilized C‐MYC activates downstream signaling to facilitate the cellular G1/S transition, proliferation, and migration. PDCD11 silencing restores SKP2‐mediated C‐MYC degradation, thereby remarkably suppressing tumor growth and metastasis in nude mice. These findings highlight PDCD11 as a novel C‐MYC partner and thereby offer a potential therapeutic rationale to challenge PDCD11‐mediated “pro‐stabilization” effect on C‐MYC, a widely considered “undruggable” target, to combat C‐MYC‐driven malignancies with p53 mutation.
Collapse
Affiliation(s)
- Li Ding
- College of Bioscience and Biotechnology Yangzhou University Yangzhou 225009 China
| | - Wei Ni
- College of Bioscience and Biotechnology Yangzhou University Yangzhou 225009 China
| | - Yichao Ma
- Department of General Surgery Institute of General Surgery Northern Jiangsu People's Hospital Affiliated to Yangzhou University Yangzhou 225001 China
| | - Lin Xu
- College of Bioscience and Biotechnology Yangzhou University Yangzhou 225009 China
| | - Zhiping Zhang
- College of Bioscience and Biotechnology Yangzhou University Yangzhou 225009 China
| | - Kai Liao
- College of Bioscience and Biotechnology Yangzhou University Yangzhou 225009 China
| | - Jingwen Li
- College of Bioscience and Biotechnology Yangzhou University Yangzhou 225009 China
| | - Xinyu Mei
- College of Bioscience and Biotechnology Yangzhou University Yangzhou 225009 China
| | - Zhun Wang
- College of Bioscience and Biotechnology Yangzhou University Yangzhou 225009 China
| | - Huiqian Ge
- College of Bioscience and Biotechnology Yangzhou University Yangzhou 225009 China
| | - Jiajia Li
- College of Bioscience and Biotechnology Yangzhou University Yangzhou 225009 China
| | - Dong Tang
- Department of General Surgery Institute of General Surgery Northern Jiangsu People's Hospital Affiliated to Yangzhou University Yangzhou 225001 China
| | - Xinyue Zhang
- College of Bioscience and Biotechnology Yangzhou University Yangzhou 225009 China
| |
Collapse
|
17
|
Liu D, Liu L, Zhang X, Zhao X, Li X, Che X, Wu G. Decoding driver and phenotypic genes in cancer: Unveiling the essence behind the phenomenon. Mol Aspects Med 2025; 103:101358. [PMID: 40037122 DOI: 10.1016/j.mam.2025.101358] [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: 10/27/2024] [Revised: 01/25/2025] [Accepted: 02/26/2025] [Indexed: 03/06/2025]
Abstract
Gray hair, widely regarded as a hallmark of aging. While gray hair is associated with aging, reversing this trait through gene targeting does not alter the fundamental biological processes of aging. Similarly, certain oncogenes (such as CXCR4, MMP-related genes, etc.) can serve as markers of tumor behavior, such as malignancy or prognosis, but targeting these genes alone may not lead to tumor regression. We pioneered the name of this class of genes as "phenotypic genes". Historically, cancer genetics research has focused on tumor driver genes, while genes influencing cancer phenotypes have been relatively overlooked. This review explores the critical distinction between driver genes and phenotypic genes in cancer, using the MAPK and PI3K/AKT/mTOR pathways as key examples. We also discuss current research techniques for identifying driver and phenotypic genes, such as whole-genome sequencing (WGS), RNA sequencing (RNA-seq), RNA interference (RNAi), CRISPR-Cas9, and other genomic screening methods, alongside the concept of synthetic lethality in driver genes. The development of these technologies will help develop personalized treatment strategies and precision medicine based on the characteristics of relevant genes. By addressing the gap in discussions on phenotypic genes, this review significantly contributes to clarifying the roles of driver and phenotypic genes, aiming at advancing the field of targeted cancer therapy.
Collapse
Affiliation(s)
- Dequan Liu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Lei Liu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Xiaoman Zhang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Xinming Zhao
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Xiaorui Li
- Department of Oncology, Cancer Hospital of Dalian University of Technology, Shenyang, 110042, China.
| | - Xiangyu Che
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China.
| | - Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China.
| |
Collapse
|
18
|
Song X, Pan Z, Zhang Y, Yang W, Zhang T, Wang H, Chen Y, Yu X, Ding H, Li R, Ge P, Xu L, Dong G, Jiang F. Excessive MYC Orchestrates Macrophages induced Chromatin Remodeling to Sustain Micropapillary-Patterned Malignancy in Lung Adenocarcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2403851. [PMID: 39899538 PMCID: PMC11948069 DOI: 10.1002/advs.202403851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 01/15/2025] [Indexed: 02/05/2025]
Abstract
Current understanding of micropapillary (MP)-subtype lung adenocarcinoma (LUAD) remains confined to biological activities and genomic landscapes. Unraveling the major regulatory programs underlying MP patterned malignancy offers opportunities to identify more feasible therapeutic targets for patients with MP LUAD. This study shows that patients with MP subtype LUAD have aberrant activation of the MYC pathway compared to patients with other subtypes. In vitro and xenograft mouse model studies reveal that MP pattern in malignancy cannot be solely due to aberrant MYC expression but requires the involvement of M2-like macrophages. Excessively expressed MYC leads to the accumulation of M2-like macrophages from the bone marrow, which secretes TGFβ, to induce the expression of FOSL2 in tumor cells, thereby remodeling chromatin accessibility at promoter regions of MP-pattern genes to promote the MYC-mediated de novo transcriptional regulation of these genes. Additionally, the MP-pattern in malignancy can be effectively alleviated by disrupting the TGFβ-FOSL2 axis. These findings reveal new functions for the M2-like macrophage-TGFβ-FOSL2 axis in MYC-overexpressing MP-subtype LUAD, identifying targetable vulnerabilities in this pathway.
Collapse
Affiliation(s)
- Xuming Song
- Department of Thoracic SurgeryNanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjing210009P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
- The Fourth Clinical College of Nanjing Medical UniversityNanjing210000P. R. China
| | - Zehao Pan
- Department of Thoracic SurgeryNanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjing210009P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
- The Fourth Clinical College of Nanjing Medical UniversityNanjing210000P. R. China
| | - Yi Zhang
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
- Department of PathologyNanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjing210009P. R. China
| | - Wenmin Yang
- Department of Thoracic SurgeryNanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjing210009P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
- Department of PathologyNanjing Drum Tower hospitalNanjing210008P.R. China
| | - Te Zhang
- Department of Thoracic SurgeryNanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjing210009P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
- Department of Biochemistry and Molecular GeneticsFeinberg School of MedicineNorthwestern UniversityChicagoIllinois60201USA
| | - Hui Wang
- Department of Thoracic SurgeryNanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjing210009P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
- The Fourth Clinical College of Nanjing Medical UniversityNanjing210000P. R. China
| | - Yuzhong Chen
- Department of Thoracic SurgeryNanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjing210009P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
- The Fourth Clinical College of Nanjing Medical UniversityNanjing210000P. R. China
| | - Xinnian Yu
- Department of Thoracic SurgeryNanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjing210009P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
- The Fourth Clinical College of Nanjing Medical UniversityNanjing210000P. R. China
| | - Hanlin Ding
- Department of Thoracic SurgeryNanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjing210009P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
- The Fourth Clinical College of Nanjing Medical UniversityNanjing210000P. R. China
| | - Rutao Li
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
- Department of Thoracic SurgeryThe Fourth Affiliated Hospital of Soochow UniversityNanjing215000P. R. China
| | - Pengfei Ge
- Department of Thoracic SurgeryNanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjing210009P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
- The Fourth Clinical College of Nanjing Medical UniversityNanjing210000P. R. China
| | - Lin Xu
- Department of Thoracic SurgeryNanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjing210009P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
- Collaborative Innovation Center for Cancer Personalized MedicineNanjing Medical UniversityNanjing211116P. R. China
| | - Gaochao Dong
- Department of Thoracic SurgeryNanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjing210009P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
| | - Feng Jiang
- Department of Thoracic SurgeryNanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer ResearchNanjing210009P. R. China
- Jiangsu Key Laboratory of Molecular and Translational Cancer ResearchCancer Institute of Jiangsu ProvinceNanjing210000P. R. China
| |
Collapse
|
19
|
Avolio E, Bassani B, Campanile M, Mohammed KA, Muti P, Bruno A, Spinetti G, Madeddu P. Shared molecular, cellular, and environmental hallmarks in cardiovascular disease and cancer: Any place for drug repurposing? Pharmacol Rev 2025; 77:100033. [PMID: 40148035 DOI: 10.1016/j.pharmr.2024.100033] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 12/17/2024] [Indexed: 03/29/2025] Open
Abstract
Cancer and cardiovascular disease (CVD) are the 2 biggest killers worldwide. Specific treatments have been developed for the 2 diseases. However, mutual therapeutic targets should be considered because of the overlap of cellular and molecular mechanisms. Cancer research has grown at a fast pace, leading to an increasing number of new mechanistic treatments. Some of these drugs could prove useful for treating CVD, which realizes the concept of cancer drug repurposing. This review provides a comprehensive outline of the shared hallmarks of cancer and CVD, primarily ischemic heart disease and heart failure. We focus on chronic inflammation, altered immune response, stromal and vascular cell activation, and underlying signaling pathways causing pathological tissue remodeling. There is an obvious scope for targeting those shared mechanisms, thereby achieving reciprocal preventive and therapeutic benefits. Major attention is devoted to illustrating the logic, advantages, challenges, and viable examples of drug repurposing and discussing the potential influence of sex, gender, age, and ethnicity in realizing this approach. Artificial intelligence will help to refine the personalized application of drug repurposing for patients with CVD. SIGNIFICANCE STATEMENT: Cancer and cardiovascular disease (CVD), the 2 biggest killers worldwide, share several underlying cellular and molecular mechanisms. So far, specific therapies have been developed to tackle the 2 diseases. However, the development of new cardiovascular drugs has been slow compared with cancer drugs. Understanding the intersection between pathological mechanisms of the 2 diseases provides the basis for repurposing cancer therapeutics for CVD treatment. This approach could allow the rapid development of new drugs for patients with CVDs.
Collapse
Affiliation(s)
- Elisa Avolio
- Bristol Heart Institute, Laboratory of Experimental Cardiovascular Medicine, Translational Health Sciences, Bristol Medical School, University of Bristol, United Kingdom.
| | - Barbara Bassani
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, IRCCS MultiMedica, Milan, Italy
| | - Marzia Campanile
- Laboratory of Cardiovascular Pathophysiology - Regenerative Medicine, IRCCS MultiMedica, Milan, Italy; Department of Biosciences, University of Milan, Milan, Italy
| | - Khaled Ak Mohammed
- Bristol Heart Institute, Laboratory of Experimental Cardiovascular Medicine, Translational Health Sciences, Bristol Medical School, University of Bristol, United Kingdom; Department of Cardiothoracic Surgery, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Paola Muti
- IRCCS MultiMedica, Milan, Italy; Department of Biomedical, Surgical and Dental Health Sciences, University of Milan, Italy
| | - Antonino Bruno
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, IRCCS MultiMedica, Milan, Italy; Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy.
| | - Gaia Spinetti
- Laboratory of Cardiovascular Pathophysiology - Regenerative Medicine, IRCCS MultiMedica, Milan, Italy.
| | - Paolo Madeddu
- Bristol Heart Institute, Laboratory of Experimental Cardiovascular Medicine, Translational Health Sciences, Bristol Medical School, University of Bristol, United Kingdom.
| |
Collapse
|
20
|
Liang Y, Mi Z, Kuo PC. Differential MYC and PROM1 mRNA isoform expression in breast invasive carcinoma as biomarkers for subtyping and prognosis. Surgery 2025; 179:108798. [PMID: 39306567 DOI: 10.1016/j.surg.2024.07.056] [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: 05/01/2024] [Revised: 06/26/2024] [Accepted: 07/14/2024] [Indexed: 02/02/2025]
Abstract
BACKGROUND Cancer stem cells are a subpopulation of tumor cells with the ability to self-renew; evidence suggests that cancer stem cells are responsible for recurrence, metastasis, and resistance to therapy. MYC and CD133 (PROM1 gene) are clinical biomarkers for cancer stem cells, and their dysregulation is involved in the progression of many cancers. Alternative splicing of these genes may contribute to cancer stem cell differentiation. METHODS Transcriptional and clinical data of PROM1 and MYC mRNA isoforms in breast cancer samples were downloaded from the TCGA Splicing Variants Database site, a web-tool to explore mRNA alternative-splicing based on TCGA samples. Data include RSEM isoform expression, clinical sample types, survival data, and clinical receptor expression. Breast cancer subtypes (luminal A, luminal B, Her2 positive, triple negative) were assigned on the basis of estrogen, progesterone, and HER2 expression. RESULTS Expression of MYC isoforms uc003ysh.1 and uc003ysi.3 was significantly greater in triple-negative breast cancer compared with all other breast cancer subtypes (P < .001). Isoform uc003ysi.3 was associated with greater 5-year survival in luminal A breast cancer (hazard ratio, 0.79; 95% confidence interval, 0.65-0.96; P = .02). PROM1 isoforms uc003gop.2, uc003goq.3, uc003gos.2, and uc003gou.2 were expressed greatest in triple-negative breast cancer (P < .001). PROM1 isoform uc003gou.2 was associated with better 5-year survival in luminal A breast cancer (hazard ratio, 0.79; 95% confidence interval, 0.65-0.97; P = .02). CONCLUSIONS MYC and PROM1 isoforms are differentially expressed in breast cancer subtypes. Certain isoforms confer better survival prognosis. Further work should be done to study alternative splicing in cancer stem cells.
Collapse
Affiliation(s)
- Yifan Liang
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL
| | - Zhiyong Mi
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL
| | - Paul C Kuo
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL; Bay Pines Veterans Affairs Health Care System, Bay Pines, FL.
| |
Collapse
|
21
|
Cao C, Li A, Xu C, Wu B, Yao L, Liu Y. Engineering artificial non-coding RNAs for targeted protein degradation. Nat Chem Biol 2025; 21:393-401. [PMID: 39215101 DOI: 10.1038/s41589-024-01719-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 08/06/2024] [Indexed: 09/04/2024]
Abstract
Targeted protein degradation has become a notable drug development strategy, but its application has been limited by the dependence on protein-based chimeras with restricted genetic manipulation capabilities. The use of long non-coding RNAs (lncRNAs) has emerged as a viable alternative, offering interactions with cellular proteins to modulate pathways and enhance degradation capabilities. Here we introduce a strategy employing artificial lncRNAs (alncRNAs) for precise targeted protein degradation. By integrating RNA aptamers and sequences from the lncRNA HOTAIR, our alncRNAs specifically target and facilitate the ubiquitination and degradation of oncogenic transcription factors and tumor-related proteins, such as c-MYC, NF-κB, ETS-1, KRAS and EGFR. These alncRNAs show potential in reducing malignant phenotypes in cells, both in vitro and in vivo, offering advantages in efficiency, adaptability and versatility. This research enhances knowledge of lncRNA-driven protein degradation and presents an effective method for targeted therapies.
Collapse
Affiliation(s)
- Congcong Cao
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Synthetic Biology Research Center, Health Science Center, Shenzhen University, Shenzhen, China
| | - Aolin Li
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Chaojie Xu
- Department of Urology, Peking University First Hospital, Beijing, China
| | - Baorui Wu
- Department of Urology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Lin Yao
- Department of Urology, Peking University First Hospital, Beijing, China.
| | - Yuchen Liu
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Synthetic Biology Research Center, Health Science Center, Shenzhen University, Shenzhen, China.
| |
Collapse
|
22
|
Ambite I, Wan MLY, Tran HT, Nazari A, Chaudhuri A, Krintel C, Gomes I, Sabari S, Ahmadi S, Carneiro ANBM, Ishac R, Haq F, Svanborg C. Multitarget mechanism of MYC inhibition by the bacterial lon protease in disease. Sci Rep 2025; 15:6778. [PMID: 40000737 PMCID: PMC11861601 DOI: 10.1038/s41598-025-88093-2] [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: 10/31/2024] [Accepted: 01/24/2025] [Indexed: 02/27/2025] Open
Abstract
Identifying specific inhibitors of the MYC oncogene has been challenging, due to off target effects associated with MYC inhibition. This study investigated how the recombinant Escherichia coli Lon protease (rLon), which targets MYC in human cells, inhibits MYC over-activation in models of infection and cancer. In silico predictions identified specific peptide domains of bacterial Lon that target MYC and the affinity of these peptides for MYC was investigated by surface plasmon resonance. The N-terminal domain of rLon was shown to interact with the C-terminal, leucine zipper domain of MYC and MAX and to prevent MYC/MAX dimerization. Furthermore, rLon targeted and degraded c-MYC in vitro and in cellular models, through the peptidase domain. In a model of kidney infection, rLon treatment prevented, c-MYC, N-MYC and L-MYC over-expression, MYC-dependent gene expression, specifically renal toxicity genes and pathology, suggesting that rLon recognizes and corrects MYC dysregulation in this disease. The findings describe a multitarget mechanism of MYC inhibition by rLon, and the combined effects achieved by the Lon domains, targeting different MYC epitopes and MYC-dependent functions, with no evidence of toxicity or detrimental effects on homeostatic MYC expression.
Collapse
Affiliation(s)
- Ines Ambite
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Klinikgatan 28, Lund, 221 84, Sweden
| | - Murphy Lam Yim Wan
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Klinikgatan 28, Lund, 221 84, Sweden
| | - Hien Thi Tran
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Klinikgatan 28, Lund, 221 84, Sweden
| | - Atefeh Nazari
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Klinikgatan 28, Lund, 221 84, Sweden
| | - Arunima Chaudhuri
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Klinikgatan 28, Lund, 221 84, Sweden
| | - Christian Krintel
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Klinikgatan 28, Lund, 221 84, Sweden
| | - Inês Gomes
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Klinikgatan 28, Lund, 221 84, Sweden
| | - Samudra Sabari
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Klinikgatan 28, Lund, 221 84, Sweden
| | - Shahram Ahmadi
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Klinikgatan 28, Lund, 221 84, Sweden
| | - António N B M Carneiro
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Klinikgatan 28, Lund, 221 84, Sweden
| | - Rita Ishac
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Klinikgatan 28, Lund, 221 84, Sweden
| | - Farhan Haq
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Klinikgatan 28, Lund, 221 84, Sweden
| | - Catharina Svanborg
- Department of Laboratory Medicine, Division of Microbiology, Immunology and Glycobiology, Lund University, Klinikgatan 28, Lund, 221 84, Sweden.
| |
Collapse
|
23
|
Xie Y, Liu F, Wu Y, Zhu Y, Jiang Y, Wu Q, Dong Z, Liu K. Inflammation in cancer: therapeutic opportunities from new insights. Mol Cancer 2025; 24:51. [PMID: 39994787 PMCID: PMC11849313 DOI: 10.1186/s12943-025-02243-8] [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: 10/10/2024] [Accepted: 01/20/2025] [Indexed: 02/26/2025] Open
Abstract
As one part of the innate immune response to external stimuli, chronic inflammation increases the risk of various cancers, and tumor-promoting inflammation is considered one of the enabling characteristics of cancer development. Recently, there has been growing evidence on the role of anti-inflammation therapy in cancer prevention and treatment. And researchers have already achieved several noteworthy outcomes. In the review, we explored the underlying mechanisms by which inflammation affects the occurrence and development of cancer. The pro- or anti-tumor effects of these inflammatory factors such as interleukin, interferon, chemokine, inflammasome, and extracellular matrix are discussed. Since FDA-approved anti-inflammation drugs like aspirin show obvious anti-tumor effects, these drugs have unique advantages due to their relatively fewer side effects with long-term use compared to chemotherapy drugs. The characteristics make them promising candidates for cancer chemoprevention. Overall, this review discusses the role of these inflammatory molecules in carcinogenesis of cancer and new inflammation molecules-directed therapeutic opportunities, ranging from cytokine inhibitors/agonists, inflammasome inhibitors, some inhibitors that have already been or are expected to be applied in clinical practice, as well as recent discoveries of the anti-tumor effect of non-steroidal anti-inflammatory drugs and steroidal anti-inflammatory drugs. The advantages and disadvantages of their application in cancer chemoprevention are also discussed.
Collapse
Affiliation(s)
- Yifei Xie
- Department of Pathology and Forensic Medicine, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
- State Key Laboratory of Metabolic Dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, 450052, China
- The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan, 450001, China
| | - Fangfang Liu
- State Key Laboratory of Metabolic Dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, 450052, China
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450007, China
- The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan, 450001, China
| | - Yunfei Wu
- State Key Laboratory of Metabolic Dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, 450052, China
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Yuer Zhu
- State Key Laboratory of Metabolic Dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, 450052, China
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Yanan Jiang
- State Key Laboratory of Metabolic Dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, 450052, China
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450007, China
- The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan, 450001, China
| | - Qiong Wu
- State Key Laboratory of Metabolic Dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, 450052, China
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450007, China
- The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan, 450001, China
| | - Zigang Dong
- State Key Laboratory of Metabolic Dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, 450052, China.
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450007, China.
- The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan, 450001, China.
| | - Kangdong Liu
- State Key Laboratory of Metabolic Dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, 450052, China.
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450007, China.
- The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan, 450001, China.
| |
Collapse
|
24
|
Sharma P, Paul K. Selective Recognition of Oncogene Promoter C-Myc G-Quadruplex: Design, Synthesis, and In Vitro Evaluation of Naphthalimide and Imidazo[1,2- a]pyrazines for Their Anticancer Activity. ACS APPLIED BIO MATERIALS 2025; 8:1377-1396. [PMID: 39844620 DOI: 10.1021/acsabm.4c01666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
c-Myc is a transcription factor that is overexpressed in most human cancers. Despite its challenging nature, we have developed a series of naphthalimide-imidazopyrazine conjugates to target c-Myc. The library of synthesized derivatives was tested for their anticancer activity against a nine-panel of cancer cell lines. Compound 8eb showed excellent cytotoxicity against all the tested cancer cell lines, with the range of growth inhibition from -98.79% to 96.62% at a single-dose concentration of 10-5 M. Further, 8eb was employed for a 5-dose assay against the same cancer cell lines, which showed efficacy at varying concentrations with an MG-MID GI50 value of 2.61 μM. Biophysical studies were performed to explore the interaction of 8eb with c-Myc Pu27 over ct-DNA, oncogene promotor Pu22, and human telomere, with a binding constant value of 1.3 × 107 M-1. Additionally, experiments were performed to get insights into the interaction mechanism between 8eb and the c-Myc oncogene promoter. A molecular docking study unveiled the stacking of the compound with G4 DNA through groove binding, where very few reports are available, with a favorable binding energy of -9.2 kcal/mol. Moreover, the pharmacokinetic study and HOMO-LUMO energy gap analysis underscored the potency of the active candidate. The compound's binding ability toward HSA was also assessed, where results suggested effective binding of the compound to HSA, revealing its potential for easy delivery to the target site. The above findings suggested that these newly synthesized candidates with potent anticancer activity offer a promising avenue as G4 DNA c-Myc stabilizers.
Collapse
Affiliation(s)
- Palak Sharma
- Department of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala 147001, India
| | - Kamaldeep Paul
- Department of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala 147001, India
| |
Collapse
|
25
|
Lai W, Liu L, Wang S, Liu Y, Chai Y. Integrated Omics Insights into Dapagliflozin Effects in Sepsis-Induced Cardiomyopathy. Biomolecules 2025; 15:286. [PMID: 40001588 PMCID: PMC11853349 DOI: 10.3390/biom15020286] [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: 12/05/2024] [Revised: 02/03/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Sepsis-induced cardiomyopathy (SIC) is a life-threatening cardiac complication of sepsis with limited therapeutic options. Dapagliflozin, a sodium-glucose cotransporter 2 (SGLT2) inhibitor, has demonstrated cardioprotective effects in heart failure, but its role in mitigating sepsis-related cardiac dysfunction remains unclear. METHODS A retrospective cohort analysis was conducted to assess the impact of pre-hospital dapagliflozin use on major adverse cardiovascular events (MACEs) and survival in patients with SIC. Additionally, a murine SIC model was established using cecal ligation and puncture (CLP) to evaluate the effects of dapagliflozin on cardiac function, histopathology, and biomarkers of myocardial injury. Transcriptomic and metabolomic profiling, combined with multi-omics integration, was employed to elucidate the molecular mechanisms underlying dapagliflozin's cardioprotective effects. RESULTS In the clinical cohort, pre-hospital dapagliflozin use was associated with a significant reduction in the risk of MACE and improved survival outcomes. In the murine SIC model, dapagliflozin restored cardiac function, reduced biomarkers of myocardial injury, and alleviated histological damage. Multi-omics analysis revealed that dapagliflozin modulates inflammatory responses, enhances autophagy, and regulates metabolic pathways such as AMPK signaling and lipid metabolism. Key regulatory genes and metabolites were identified, providing mechanistic insights into the underlying actions of dapagliflozin. CONCLUSIONS Dapagliflozin significantly improves cardiac outcomes in sepsis-induced cardiomyopathy through the multi-level regulation of inflammation, energy metabolism, and cellular survival pathways. These findings establish dapagliflozin as a promising therapeutic strategy for SIC, offering translational insights into the treatment of sepsis-induced cardiac dysfunction.
Collapse
Affiliation(s)
| | | | | | - Yancun Liu
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yanfen Chai
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
26
|
Tang M, Crown J, Duffy MJ. Targeting MYC for the treatment of breast cancer: use of the novel MYC-GSPT1 degrader, GT19630. Invest New Drugs 2025; 43:167-179. [PMID: 39875774 PMCID: PMC11868176 DOI: 10.1007/s10637-024-01504-5] [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: 10/27/2024] [Accepted: 12/30/2024] [Indexed: 01/30/2025]
Abstract
BACKGROUND Since MYC is one of the most frequently altered driver genes involved in cancer formation, it is a potential target for new anti-cancer therapies. Historically, however, MYC has proved difficult to target due to the absence of a suitable crevice for binding potential low molecular weight drugs. OBJECTIVE The aim of this study was to evaluate a novel molecular glue, dubbed GT19630, which degrades both MYC and GSPT1, for the treatment of breast cancer. METHODS The antiproliferative potential of GT19630 was evaluated in 14 breast cancer cell lines representing the main molecular subtypes of breast cancer. In addition, we also investigated the effects of GT19630 on apoptosis, cell cycle progression, cell migration, and degradation of the negative immune checkpoint protein, B7-H3. RESULTS GT19630 inhibited cell proliferation, blocked cell cycle progression, promoted apoptosis, and decreased cell migration at low nanomolar concentrations in breast cancer cell lines. By contrast, previously described MYC inhibitors such as specific MYC-MAX antagonists affected these processes at micromolar concentrations. Consistent with the ability of MYC to promote immune evasion, we also found that GT19630 degraded the negative immune checkpoint inhibitor, B7-H3. CONCLUSIONS We conclude that the novel molecular glue, GT19630, is a potent mediator of endpoints associated with cancer formation/progression. Its ability to degrade B7-H3 suggests that GT19630 may also promote host immunity against cancer. To progress GT19630 as a therapy for breast cancer, our finding should now be confirmed in an animal model system.
Collapse
Affiliation(s)
- Minhong Tang
- UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - John Crown
- Department of Medical Oncology, St Vincent's University Hospital, Dublin, Ireland
| | - Michael J Duffy
- UCD School of Medicine, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland.
- Clinical Research Centre, St Vincent's University Hospital, Dublin, D04T6F4, Ireland.
| |
Collapse
|
27
|
Yu XX, Liu Y, Luo RJ, Song ZX, Chen WK, Mo ZM, Wang FJ. Bioinformatics analysis combined with experimental validation reveals the biological role of the ILK gene in prostate cancer. Discov Oncol 2025; 16:106. [PMID: 39890647 PMCID: PMC11785868 DOI: 10.1007/s12672-025-01852-5] [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: 08/18/2024] [Accepted: 01/24/2025] [Indexed: 02/03/2025] Open
Abstract
BACKGROUND Prostate cancer (PCa) is a prevalent urological malignancy. The integrin-linked kinase (ILK) gene has been identified as an oncogenic driver in hormonal cancers, including PCa. METHODS To identify key genes in PCa, we utilized differential gene expression analysis and Weighted Gene Co-expression Network Analysis (WGCNA). The ILK gene was silenced using short interfering RNA (siRNA), and subsequent experiments focusing on cellular functionality were conducted to evaluate its impact on cell proliferation, apoptosis, and cell cycle. We examined the expression of autophagy-related and cell cycle-related proteins, including MAP1LC3A, BECN1, C-MYC, TP53, and MDM2. Moreover, we conducted Mfuzz expression pattern clustering analysis, gene set enrichment analysis (GSEA), immune function analysis, transcription factor (TF) analysis, and drug prediction. RESULTS 544 significant genes were identified by WGCNA. The protein-protein interaction (PPI) network analysis revealed that MYC was the central regulatory gene, with the intersected genes mainly involved in regulating cell adhesion and drug metabolism in prostate cancer (PCa). Experimental results showed LNCaP cell proliferation was significantly inhibited in the knockdown groups (P < 0.001). Moreover, ILK silencing increased apoptosis in LNCaP cells compared to normal cells and empty vectors, and transfected LNCaP cells were arrested in the S phase of the cell cycle. Notably, C-MYC expression decreased following ILK silencing. Subsequently, we further identified ILK-related regulatory biomarkers. CONCLUSIONS The ILK is an oncogene mainly through influencing the C-MYC in PCa. Inhibition of ILK expression would be a promising method for treating the development and progression of PCa.
Collapse
Affiliation(s)
- Xiao-Xiang Yu
- Department of Urology, The 923, Hospital of Chinese People's Liberation Army, Nanning, 530021, Guangxi, China.
| | - Yi Liu
- Department of Urology, The 923, Hospital of Chinese People's Liberation Army, Nanning, 530021, Guangxi, China
| | - Rong-Jiang Luo
- Department of Urology, The 923, Hospital of Chinese People's Liberation Army, Nanning, 530021, Guangxi, China
| | - Zi-Xuan Song
- Department of Urology, The 923, Hospital of Chinese People's Liberation Army, Nanning, 530021, Guangxi, China
| | - Wen-Kai Chen
- Department of Urology, The 923, Hospital of Chinese People's Liberation Army, Nanning, 530021, Guangxi, China
| | - Zeng-Mi Mo
- Department of Urology, The 923, Hospital of Chinese People's Liberation Army, Nanning, 530021, Guangxi, China
| | - Feng-Jing Wang
- Department of Urology, The 923, Hospital of Chinese People's Liberation Army, Nanning, 530021, Guangxi, China
| |
Collapse
|
28
|
Lei J, Chen J, Yu W, Wu Q, Jing S, Tang Y, Lin L, Hu M. Portrait of WWP1: the current state in human cancer. Front Cell Dev Biol 2025; 12:1516613. [PMID: 39949609 PMCID: PMC11821962 DOI: 10.3389/fcell.2024.1516613] [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: 10/24/2024] [Accepted: 12/31/2024] [Indexed: 02/16/2025] Open
Abstract
WWP1, a member of the C2-WW-HECT E3 ligase family, is an E3 ubiquitin-protein ligase containing WW domains. This enzyme plays a critical role in regulating diverse cellular processes. Its expression is modulated by various factors and non-coding RNAs, resulting in ubiquitination that affects substrate protein degradation. WWP1 demonstrates a dual function, acting predominantly as an oncogene in tumors but occasionally as a tumor suppressor. This review summarizes WWP1's biological roles, therapeutic potential in oncology, upstream regulatory factors, and downstream substrates. It aims to promote research on WWP1's antitumor effects, improve understanding of its role in tumorigenesis, and support the development of targeted therapies.
Collapse
Affiliation(s)
- Jiaming Lei
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Jun Chen
- The Central Hospital of Ezhou, Affiliated Hospital of Hubei University of Science and Technology, Ezhou, Hubei, China
| | - Wenwen Yu
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Qing Wu
- State Key Laboratory of Genetic Resources and Evolution, Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Key Laboratory of Healthy Aging Study, KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Shuang Jing
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Yuanguang Tang
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Li Lin
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Meichun Hu
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China
| |
Collapse
|
29
|
Zhang D, Lu B, Ma Q, Xu W, Zhang Q, Xiao Z, Li Y, Chen R, Wang AJ. Identification of a novel immunogenic cell death-related classifier to predict prognosis and optimize precision treatment in hepatocellular carcinoma. Heliyon 2025; 11:e41380. [PMID: 39897773 PMCID: PMC11786863 DOI: 10.1016/j.heliyon.2024.e41380] [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: 03/23/2024] [Revised: 12/18/2024] [Accepted: 12/18/2024] [Indexed: 02/04/2025] Open
Abstract
Accumulating studies have highlighted the biological significance of immunogenic cell death (ICD) in cancer immunity. However, the influence of ICD on tumor microenvironment (TME) formation and immune response in Hepatocellular carcinoma (HCC) remains largely unexplored. In this study, we systematically analyzed the mRNA profiles of ICD-related genes in 1847 HCC patients and identified three molecular subtypes with significantly different immune features and prognostic stratification. A reliable risk model named ICD score was constructed via machine learning algorithms to assess the immunological status, therapeutic responses, and clinical outcomes of individual HCC patients. High ICD score indicated an immune-excluded TME phenotype, with lower anticancer immunity and shorter survival time. In contrast, low ICD score corresponded to abundant immune cell infiltration, high sensitivity to immunotherapy and a positive prognosis, indicating an "immune-hot" phenotype. Pan-cancer analysis further validated a negative association between ICD score and the immune cell infiltration levels. In conclusion, our findings revealed that the ICD score could serve as a robust prognostic biomarker to predict the benefits of immunotherapy and optimize the clinical decision-making of HCC patients.
Collapse
Affiliation(s)
- Dongjing Zhang
- Department of Gastroenterology and Hepatology, Shenzhen Clinical Research Center for Digestive Disease, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Bingyun Lu
- Department of Gastroenterology and Hepatology, Shenzhen Clinical Research Center for Digestive Disease, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Qianqian Ma
- Department of Infectious Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Wen Xu
- Department of Gastroenterology and Hepatology, Shenzhen Clinical Research Center for Digestive Disease, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Qi Zhang
- Department of Gastroenterology and Hepatology, Shenzhen Clinical Research Center for Digestive Disease, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Zhiqi Xiao
- Department of Gastroenterology and Hepatology, Shenzhen Clinical Research Center for Digestive Disease, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Yuanheng Li
- Queen Mary School, Nanchang University, Nanchang, Jiangxi, China
| | - Ren Chen
- Department of Infectious Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - An-jiang Wang
- Department of Gastroenterology and Hepatology, Shenzhen Clinical Research Center for Digestive Disease, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
| |
Collapse
|
30
|
Zhang D, Jiang M, Li P, Laster KV, Zhao D, Zhi Y, Wei H, Nie W, Gao Y, Wu Q, Xiang P, He X, Liu K, Dong Z. CHI-KAT8i5 suppresses ESCC tumor growth by inhibiting KAT8-mediated c-Myc stability. Cell Rep 2025; 44:115135. [PMID: 39772391 DOI: 10.1016/j.celrep.2024.115135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 10/15/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
The integrated analysis of histone modifier enzymes in solid tumors, especially in esophageal squamous cell carcinoma (ESCC), is still inadequate. Here, we investigate the expression levels of histone modifier enzymes in ESCC tissues. Notably, KAT8 (lysine acetyltransferase 8) is identified as a prognostic and therapeutic biomarker in ESCC. Esophageal-tissue-specific deletion of KAT8 in mice led to less tumor burden after induction of tumorigenesis via 4-nitroquinoline N-oxide (4NQO) treatment compared with wild-type mice. Meanwhile, silencing KAT8 significantly suppresses tumor growth in cell-line-derived xenograft (CDX) and patient-derived xenograft (PDX) models. Mechanically, we confirm that KAT8 regulates c-Myc protein stability by directly binding it. Furthermore, we design and screen a specific KAT8 inhibitor (CHI-KAT8i5) that significantly attenuates tumor growth in vitro and in vivo, providing promising potential for clinical application. Thus, our work identifies that KAT8 could serve as a potential clinically relevant biomarker and therapeutic target in patients with ESCC and that KAT8 inhibitor is a promising lead candidate for ESCC therapy.
Collapse
Affiliation(s)
- Dandan Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000 Henan, China; China-US (Henan) Hormel Cancer Institute, No. 127, Zhengzhou 450000 Henan, China
| | - Ming Jiang
- China-US (Henan) Hormel Cancer Institute, No. 127, Zhengzhou 450000 Henan, China
| | - Pan Li
- China-US (Henan) Hormel Cancer Institute, No. 127, Zhengzhou 450000 Henan, China
| | - Kyle Vaughn Laster
- China-US (Henan) Hormel Cancer Institute, No. 127, Zhengzhou 450000 Henan, China
| | - Dengyun Zhao
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000 Henan, China; China-US (Henan) Hormel Cancer Institute, No. 127, Zhengzhou 450000 Henan, China
| | - Yafei Zhi
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000 Henan, China; China-US (Henan) Hormel Cancer Institute, No. 127, Zhengzhou 450000 Henan, China
| | - Huifang Wei
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000 Henan, China; China-US (Henan) Hormel Cancer Institute, No. 127, Zhengzhou 450000 Henan, China
| | - Wenna Nie
- China-US (Henan) Hormel Cancer Institute, No. 127, Zhengzhou 450000 Henan, China
| | - Yunfeng Gao
- China-US (Henan) Hormel Cancer Institute, No. 127, Zhengzhou 450000 Henan, China
| | - Qiong Wu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000 Henan, China; China-US (Henan) Hormel Cancer Institute, No. 127, Zhengzhou 450000 Henan, China
| | - Pu Xiang
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450000 Henan, China
| | - Xinyu He
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000 Henan, China; China-US (Henan) Hormel Cancer Institute, No. 127, Zhengzhou 450000 Henan, China
| | - Kangdong Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000 Henan, China; China-US (Henan) Hormel Cancer Institute, No. 127, Zhengzhou 450000 Henan, China; The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou 450000 Henan, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou 450000 Henan, China.
| | - Zigang Dong
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000 Henan, China; China-US (Henan) Hormel Cancer Institute, No. 127, Zhengzhou 450000 Henan, China; The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou 450000 Henan, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou 450000 Henan, China.
| |
Collapse
|
31
|
Ließem A, Leimer U, Germann GK, Köllensperger E. Adipokines in Breast Cancer: Decoding Genetic and Proteomic Mechanisms Underlying Migration, Invasion, and Proliferation. BREAST CANCER (DOVE MEDICAL PRESS) 2025; 17:79-102. [PMID: 39882382 PMCID: PMC11776935 DOI: 10.2147/bctt.s491277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 12/04/2024] [Indexed: 01/31/2025]
Abstract
Background Adipokines, bioactive peptides secreted by adipose tissue, appear to contribute to breast cancer development and progression. While numerous studies suggest their role in promoting tumor growth, the exact mechanisms of their involvement are not yet completely understood. Methods In this project, varying concentrations of recombinant human adipokines (Leptin, Lipocalin-2, PAI-1, and Resistin) were used to study their effects on four selected breast cancer cell lines (EVSA-T, MCF-7, MDA-MB-231, and SK-Br-3). Over a five-day proliferation phase, linear growth was assessed by calculating doubling times and malignancy-associated changes in gene and protein expression were identified using quantitative TaqMan real-time PCR and multiplex protein analysis. Migration and invasion behaviors were quantified using specialized Boyden chamber assays. Results We found significant, adipokine-mediated genetic and proteomic alterations, with PCR showing an up to 6-fold increase of numerous malignancy-associated genes after adipokine-supplementation. Adipokines further altered protein secretion, such as raising the concentrations of different tumor-associated proteins up to 13-fold. Effects on proliferation varied, however, with most approaches showing significant enhancement in growth kinetics. A concentration-dependent increase in migration and invasion was generally observed, with no significant reductions in any approaches. Conclusion We could show a robust promoting effect of several adipokines on different breast cancer cells in vitro. Understanding the interaction between adipose tissue and breast cancer cells opens potential avenues for innovative breast cancer prevention and therapy strategies. Our findings indicate that antibodies against specific adipokines could become a beneficial component of clinical breast cancer treatment in the future.
Collapse
Affiliation(s)
- Anne Ließem
- Clinic for Plastic, Aesthetic and Reconstructive Surgery, Spine, Orthopedic and Hand Surgery, Preventive Medicine – ETHIANUM, Heidelberg, 69115, Germany
| | - Uwe Leimer
- Clinic for Plastic, Aesthetic and Reconstructive Surgery, Spine, Orthopedic and Hand Surgery, Preventive Medicine – ETHIANUM, Heidelberg, 69115, Germany
| | - Günter K Germann
- Clinic for Plastic, Aesthetic and Reconstructive Surgery, Spine, Orthopedic and Hand Surgery, Preventive Medicine – ETHIANUM, Heidelberg, 69115, Germany
| | - Eva Köllensperger
- Clinic for Plastic, Aesthetic and Reconstructive Surgery, Spine, Orthopedic and Hand Surgery, Preventive Medicine – ETHIANUM, Heidelberg, 69115, Germany
| |
Collapse
|
32
|
Li C, Sun Y. Investigation of chromatin remodeling-related biomarkers and associated molecular mechanism in pulpitis. Gene 2025; 934:149016. [PMID: 39433267 DOI: 10.1016/j.gene.2024.149016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 09/26/2024] [Accepted: 10/16/2024] [Indexed: 10/23/2024]
Abstract
The current study aimed to identify potential chromatin remodeling-related biomarkers and the associated molecular mechanisms in pulpitis. Differentially expressed genes associated with chromatin remodeling (DECRGs) were identified using datasets from an online database. Enrichment and protein-protein interaction (PPI) network analyses were performed based on the DECRGs to identify biomarkers for pulpitis, followed by GSEA (gene set enrichment analysis). The diagnostic value of these biomarkers were evaluated by ROC (Receiver operating characteristic) and nomogram investigation. Next, microRNA(miRNA)-mRNA-TF (transcription factor), ceRNA (competing endogenous RNA), and drug prediction networks were constructed based on the biomarkers. Finally, reverse transcription-real-time quantitative PCR analysis and western blot were performed to validate the results of the bioinformatic analysis. This study identified 87 DECRGs between pulpitis and normal dental pulp samples that were mainly enriched in chromatin remodeling functions and pathways in cancer. A PPI network identified five biomarkers: TNF, STAT3, MYC, ACTB, and MAPK8. ROC and nomogram analyses demonstrated the diagnostic value of these biomarkers. GSEA of biomarkers such as STAT3 was mainly enriched in functions such as the B cell receptor signaling pathway. A biomarker-disease network and miRNA-mRNA-TF interactions were constructed using these biomarkers. A ceRNA network was constructed with interactions including chr22-38_28785274-29006793.1-miR-125b-5p-STAT3. A drug-gene network was established using 170 drugs and five biomarkers. Finally, qRT-PCR was used to validate the expression of all five biomarkers identified by the bioinformatics analysis. TNF, STAT3, MYC, ACTB, and MAPK8 are potential chromatin remodeling-related diagnostic markers for pulpitis. Moreover, long non-coding RNA (lncRNA) chr22-38_28785274-29006793.1 might function as a ceRNA to regulate the expression of the chromatin remodeling gene STAT3 by sponging miR-125b-5p in pulpitis.
Collapse
Affiliation(s)
- Chenglin Li
- Department of Stomatology, Sunshine Union Hospital, Weifang 261000, China
| | - Yujiao Sun
- Department of Stomatology, Sunshine Union Hospital, Weifang 261000, China.
| |
Collapse
|
33
|
Okun SA, Lu D, Sew K, Subramaniam A, Lockwood WW. MET Activation in Lung Cancer and Response to Targeted Therapies. Cancers (Basel) 2025; 17:281. [PMID: 39858062 PMCID: PMC11764361 DOI: 10.3390/cancers17020281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/09/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
The hepatocyte growth factor receptor (MET) is a receptor tyrosine kinase (RTK) that mediates the activity of a variety of downstream pathways upon its activation. These pathways regulate various physiological processes within the cell, including growth, survival, proliferation, and motility. Under normal physiological conditions, this allows MET to regulate various development and regenerative processes; however, mutations resulting in aberrant MET activity and the consequent dysregulation of downstream signaling can contribute to cellular pathophysiology. Mutations within MET have been identified in a variety of cancers and have been shown to mediate tumorigenesis by increasing RTK activity and downstream signaling. In lung cancer specifically, a number of patients have been identified as possessing MET alterations, commonly receptor amplification (METamp) or splice site mutations resulting in loss of exon 14 (METex14). Due to MET's role in mediating oncogenesis, it has become an attractive clinical target and has led to the development of various targeted therapies, including MET tyrosine kinase inhibitors (TKIs). Unfortunately, these TKIs have demonstrated limited clinical efficacy, as patients often present with either primary or acquired resistance to these therapies. Mechanisms of resistance vary but often occur through off-target or bypass mechanisms that render downstream signaling pathways insensitive to MET inhibition. This review provides an overview of the therapeutic landscape for MET-positive cancers and explores the various mechanisms that contribute to therapeutic resistance in these cases.
Collapse
Affiliation(s)
- Sarah Anna Okun
- Integrative Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (S.A.O.); (K.S.); (A.S.)
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Daniel Lu
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Katherine Sew
- Integrative Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (S.A.O.); (K.S.); (A.S.)
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Asha Subramaniam
- Integrative Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (S.A.O.); (K.S.); (A.S.)
- Department of Pathology and Laboratory Science, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - William W. Lockwood
- Integrative Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (S.A.O.); (K.S.); (A.S.)
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Department of Pathology and Laboratory Science, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
34
|
Guo X, Feng C, Xing J, Cao Y, Liu T, Yang W, Mu R, Wang T. Epigenetic profiling for prognostic stratification and personalized therapy in breast cancer. Front Immunol 2025; 15:1510829. [PMID: 39877345 PMCID: PMC11772270 DOI: 10.3389/fimmu.2024.1510829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 12/16/2024] [Indexed: 01/31/2025] Open
Abstract
Background The rising incidence of breast cancer and its heterogeneity necessitate precise tools for predicting patient prognosis and tailoring personalized treatments. Epigenetic changes play a critical role in breast cancer progression and therapy responses, providing a foundation for prognostic model development. Methods We developed the Machine Learning-derived Epigenetic Model (MLEM) to identify prognostic epigenetic gene patterns in breast cancer. Using multi-cohort transcriptomic datasets, MLEM was constructed with rigorous machine learning techniques and validated across independent datasets. The model's performance was further corroborated through immunohistochemical validation on clinical samples. Results MLEM effectively stratified breast cancer patients into high- and low-risk groups. Low-MLEM patients exhibited improved prognosis, characterized by enhanced immune cell infiltration and higher responsiveness to immunotherapy. High-MLEM patients showed poorer prognosis but were more responsive to chemotherapy, with vincristine identified as a promising therapeutic option. The model demonstrated robust performance across independent validation datasets. Conclusion MLEM is a powerful prognostic tool for predicting breast cancer outcomes and tailoring personalized treatments. By integrating epigenetic insights with machine learning, this model has the potential to improve clinical decision-making and optimize therapeutic strategies for breast cancer patients.
Collapse
Affiliation(s)
- Xiao Guo
- School of Pharmacy, Beihua University, Jilin, China
| | - Chuanbo Feng
- School of Pharmacy, Beihua University, Jilin, China
| | - Jiaying Xing
- School of Pharmacy, Beihua University, Jilin, China
| | - Yuyan Cao
- School of Pharmacy, Beihua University, Jilin, China
| | - Tengda Liu
- School of Pharmacy, Beihua University, Jilin, China
| | | | - Runhong Mu
- School of Basic Medical Sciences, Beihua University, Jilin, China
| | - Tao Wang
- Research Laboratory Center, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, China
| |
Collapse
|
35
|
Seres M, Spacayova K, Sulova Z, Spaldova J, Breier A, Pavlikova L. Dynamic Multilevel Regulation of EGFR, KRAS, and MYC Oncogenes: Driving Cancer Cell Proliferation Through (Epi)Genetic and Post-Transcriptional/Translational Pathways. Cancers (Basel) 2025; 17:248. [PMID: 39858030 PMCID: PMC11763799 DOI: 10.3390/cancers17020248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
The epidermal growth factor receptor (EGFR) regulates gene expression through two primary mechanisms: as a growth factor in the nucleus, where it translocates upon binding its ligand, or via its intrinsic tyrosine kinase activity in the cytosol, where it modulates key signaling pathways such as RAS/MYC, PI3K, PLCγ, and STAT3. During tumorigenesis, these pathways become deregulated, leading to uncontrolled proliferation, enhanced migratory and metastatic capabilities, evasion of programmed cell death, and resistance to chemotherapy or radiotherapy. The RAS and MYC oncogenes are pivotal in tumorigenesis, driving processes such as resistance to apoptosis, replicative immortality, cellular invasion and metastasis, and metabolic reprogramming. These oncogenes are subject to regulation by a range of epigenetic and post-transcriptional modifications. This review focuses on the deregulation of EGFR, RAS, and MYC expression caused by (epi)genetic alterations and post-translational modifications. It also explores the therapeutic potential of targeting these regulatory proteins, emphasizing the importance of phenotyping neoplastic tissues to inform the treatment of cancer.
Collapse
Affiliation(s)
- Mario Seres
- Institute of Molecular Physiology and Genetics, Centre of Bioscience, Slovak Academy of Sciences, Dúbravská Cesta 9, 84005 Bratislava, Slovakia; (M.S.); (K.S.); (Z.S.)
| | - Katarina Spacayova
- Institute of Molecular Physiology and Genetics, Centre of Bioscience, Slovak Academy of Sciences, Dúbravská Cesta 9, 84005 Bratislava, Slovakia; (M.S.); (K.S.); (Z.S.)
- Department of Molecular Biology, Faculty of Natural Sciences, Comenius University, Ilkovičova 6, 84215 Bratislava, Slovakia
| | - Zdena Sulova
- Institute of Molecular Physiology and Genetics, Centre of Bioscience, Slovak Academy of Sciences, Dúbravská Cesta 9, 84005 Bratislava, Slovakia; (M.S.); (K.S.); (Z.S.)
| | - Jana Spaldova
- Institute of Biochemistry and Microbiology, Faculty of Chemical and Food Technology, Slovak University of Technology, Radlinského 9, 81237 Bratislava, Slovakia;
| | - Albert Breier
- Institute of Molecular Physiology and Genetics, Centre of Bioscience, Slovak Academy of Sciences, Dúbravská Cesta 9, 84005 Bratislava, Slovakia; (M.S.); (K.S.); (Z.S.)
- Institute of Biochemistry and Microbiology, Faculty of Chemical and Food Technology, Slovak University of Technology, Radlinského 9, 81237 Bratislava, Slovakia;
| | - Lucia Pavlikova
- Institute of Molecular Physiology and Genetics, Centre of Bioscience, Slovak Academy of Sciences, Dúbravská Cesta 9, 84005 Bratislava, Slovakia; (M.S.); (K.S.); (Z.S.)
| |
Collapse
|
36
|
Yao K, Fan H, Yang T, Yang C, Wang G, Li X, Ji XY, Wang Q, Lv S, Guo S. Identification of MYC and STAT3 for early diagnosis based on the long noncoding RNA-mRNA network and bioinformatics in colorectal cancer. Front Immunol 2025; 15:1497919. [PMID: 39830506 PMCID: PMC11739134 DOI: 10.3389/fimmu.2024.1497919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 12/05/2024] [Indexed: 01/22/2025] Open
Abstract
Background Colorectal cancer (CRC) ranks among the top three cancers globally in both incidence and mortality, posing a significant public health challenge. Most CRC cases are diagnosed at intermediate to advanced stages, and reliable biomarkers for early detection are lacking. Long non-coding RNAs (lncRNAs) have been implicated in various cancers, including CRC, playing key roles in tumor development, progression, and prognosis. Methods A comprehensive search of the PubMed database was conducted to identify relevant studies on the early diagnosis of CRC. Bioinformatics analysis was performed to explore lncRNA-mRNA networks, leading to the identification of five potential blood biomarkers. Expression analysis was carried out using the GEPIA and GEO online databases, focusing on MYC and STAT3. Differential expression between normal and CRC tissues was assessed, followed by Receiver Operating Characteristic (ROC) analysis to evaluate the diagnostic potential of these markers. Quantitative Real-Time PCR (qRT-PCR) was performed to validate MYC and STAT3 expression levels, and findings were further confirmed using the Human Protein Atlas (HPA) database. Results Database analysis revealed significant differential expression of MYC and STAT3 between normal and CRC tissues. ROC analysis demonstrated the diagnostic potential of these markers. qRT-PCR validation confirmed the differential expression patterns observed in the databases. Validation through the HPA database further supported these findings, confirming the potential of MYC and STAT3 as diagnostic biomarkers for CRC. Conclusion Our results suggest that MYC and STAT3 are promising diagnostic biomarkers for CRC, offering new insights into its pathophysiology and potential for targeted therapies.
Collapse
Affiliation(s)
- Kunhou Yao
- Department of General Surgery, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan, China
| | - Hao Fan
- School of Basic Medicine, Henan University, Kaifeng, Henan, China
| | - Tiancheng Yang
- School of Basic Medicine, Henan University, Kaifeng, Henan, China
| | - Can Yang
- School of Basic Medicine, Henan University, Kaifeng, Henan, China
| | - Guibin Wang
- School of Basic Medicine, Henan University, Kaifeng, Henan, China
| | - Xingwang Li
- Department of General Surgery, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan, China
| | - Xin-Ying Ji
- Department of General Surgery, Huaxian County People’s Hospital, Huaxian, Henan, China
| | - Qun Wang
- School of Basic Medicine, Henan University, Kaifeng, Henan, China
| | - Shaojiang Lv
- Department of General Surgery, Huaxian County People’s Hospital, Huaxian, Henan, China
| | - Shihao Guo
- Department of Colorectal Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
37
|
Liu Z, Jiang H, Kan H, Zhang L, Rao Y, Jiang X, Li M, Wang Q. RIT1 Promotes the Proliferation of Gliomas Through the Regulation of the PI3K/AKT/c-Myc Signalling Pathway. J Cell Mol Med 2025; 29:e70362. [PMID: 39833023 PMCID: PMC11745823 DOI: 10.1111/jcmm.70362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/28/2024] [Accepted: 01/06/2025] [Indexed: 01/22/2025] Open
Abstract
Recently, RIT1 has been implicated in a range of neurological disorders; however, its precise function in glioma pathogenesis is not yet well-defined. This study employed quantitative reverse transcription PCR (qRT-PCR), Western blotting (WB), immunohistochemistry (IHC) and additional methodologies to assess RIT1 expression levels in glioma tissues. Furthermore, the study investigated its influence on glioma progression through a series of functional experiments. Animal models were also utilised to elucidate the mechanistic role of RIT1, with a particular focus on its effects on the PI3K/AKT signalling pathway. Research findings showcased that RIT1 is significantly overexpressed in gliomas and exhibits a strong correlation with tumour grade and unfavourable clinical outcomes. Furthermore, RIT1 serves as an independent prognostic marker of poor prognosis. Functional assays demonstrate that RIT1 facilitates the aggressiveness of glioma cells by activating the PI3K/AKT signalling. Additionally, it promotes tumour proliferation by inhibiting apoptosis and accelerating cell cycle progression. This study demonstrates that RIT1 significantly contributes to the aggressive phenotype and unfavourable prognosis of glioma, indicating its ability as a therapeutic target for glioma treatment.
Collapse
Affiliation(s)
- Zhen Liu
- Department of Neurosurgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| | - Hao‐dong Jiang
- Department of Neurosurgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| | - Hao‐yuan Kan
- Department of Neurosurgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| | - Li Zhang
- Department of Neurosurgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| | - Yu‐xin Rao
- Department of Neurosurgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| | - Xiao‐bing Jiang
- Department of Neurosurgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| | - Ming‐hui Li
- Department of AnesthesiologyHubei University of Chinese Medicine Affiliated Hubei Hospital of Chinese MedicineWuhanChina
| | - Qi Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology, Ministry of EducationWuhanChina
| |
Collapse
|
38
|
Camps-Fajol C, Cavero D, Minguillón J, Surrallés J. Targeting protein-protein interactions in drug discovery: Modulators approved or in clinical trials for cancer treatment. Pharmacol Res 2025; 211:107544. [PMID: 39667542 DOI: 10.1016/j.phrs.2024.107544] [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: 10/16/2024] [Revised: 11/27/2024] [Accepted: 12/09/2024] [Indexed: 12/14/2024]
Abstract
Protein-protein interactions (PPIs) form complex cellular networks fundamental to many key biological processes, including signal transduction, cell proliferation and DNA repair. In consequence, their perturbation is often associated with many human diseases. Targeting PPIs offers a promising approach in drug discovery and ongoing advancements in this field hold the potential to provide highly specific therapies for a wide range of complex diseases. Despite the development of PPI modulators is challenging, advances in the genetic, proteomic and computational level have facilitated their discovery and optimization. Focusing on anticancer drugs, in the last years several PPI modulators have entered clinical trials and venetoclax, which targets Bcl-2 family proteins, has been approved for treating different types of leukemia. This review discusses the clinical development status of drugs modulating several PPIs, such as MDM2-4/p53, Hsp90/Hsp90, Hsp90/CDC37, c-Myc/Max, KRAS/SOS1, CCR5/CCL5, CCR2/CCL2 or Smac/XIAP, in cancer drug discovery.
Collapse
Affiliation(s)
- Cristina Camps-Fajol
- Unitat Mixta de Recerca en Medicina Genòmica, Universitat Autònoma de Barcelona (UAB)-IR SANT PAU, Barcelona, Spain; Institut de Bioenginyeria de Catalunya (IBEC), Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III (CIBERER, ISCIII), Madrid, Spain
| | - Debora Cavero
- Unitat Mixta de Recerca en Medicina Genòmica, Universitat Autònoma de Barcelona (UAB)-IR SANT PAU, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III (CIBERER, ISCIII), Madrid, Spain
| | - Jordi Minguillón
- CIBERER-ISCIII, IdiPAZ-CNIO Translational Research Unit in Pediatric Hemato-Oncology, La Paz University Hospital Research Institute; Spanish National Cancer Center, Madrid, Spain; Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
| | - Jordi Surrallés
- Unitat Mixta de Recerca en Medicina Genòmica, Universitat Autònoma de Barcelona (UAB)-IR SANT PAU, Barcelona, Spain; Institut de Bioenginyeria de Catalunya (IBEC), Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III (CIBERER, ISCIII), Madrid, Spain; Servei de Genètica, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Departament de Genètica i Microbiologia, Universitat Autònoma de Barcelona, Spain.
| |
Collapse
|
39
|
Xie T, Gao Y, Hu J, Luo R, Guo Y, Xie Q, Yan C, Tang Y, Chen P, Yang Z, Yu Q, Hu F, Zhang X. Increased matrix stiffness in pituitary neuroendocrine tumors invading the cavernous sinus is activated by TAFs: focus on the mechanical signatures. Endocrine 2025; 87:281-294. [PMID: 39240459 DOI: 10.1007/s12020-024-04022-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 08/27/2024] [Indexed: 09/07/2024]
Abstract
PURPOSE Pituitary neuroendocrine tumors (PitNETs) with invasion of the cavernous sinus (CS) are particularly challenging to treat. Tumor associated fibroblasts (TAFs) are recognized for their pivotal role in reprogramming extracellular matrix (ECM). Herein, we aimed to explore the potential involvement of TAFs in ECM reprogramming and elucidate the underlying mechanism involved. METHODS We applied dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) to measure tumor vessel permeability and applied atomic force microscopy (AFM) to measure the matrix stiffness of PitNETs located in both CS and sella turcica (ST). Western blotting, immunofluorescence, immunohistochemistry, and quantitative RT-PCR were utilized to analyze the ECM components. Proteomic biochemical analysis was utilized to uncover potential mechanisms governing ECM dynamics. RESULTS We found that PitNETs in the CS were stiffer than those in the ST. Increased ECM stiffness within the CS facilitated the acquisition of stem-like properties, enhanced proliferation, and induced epithelial-to-mesenchymal transition (EMT) of GH3 cells. Furthermore, the expression levels of lysyl oxidase (LOX), matrix metallopeptidase 2 (MMP2) and MMP9 in pituitary adenoma cells increased in the stiffer matrix. Proteomic analysis suggested TAFs were activated in the CS area and contributed enhanced matrix stiffness by secreting Col-1 and Col-3. Furthermore, mTOR pathway was activated under higher matrix stiffness and the migration and invasion of GH3 cells be repressed by mTOR inhibitor. CONCLUSION These findings demonstrated that activated TAFs contributed to stiffer matrix and increased ECM stiffness stimulating mTOR pathway in pituitary tumor cells. Our study indicated that mTOR inhibitor was a promising treatment strategy from the standpoint of PitNET biomechanical properties.
Collapse
Affiliation(s)
- Tao Xie
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China
- Department of Neurosurgery, Shanghai Geriatric Medical Center, 2560 Chunsheng Road, Shanghai, China
- Cancer Center, Shanghai Zhongshan Hospital, Fudan University, Shanghai, China
- The innovation and translation alliance of neuroendoscopy in the Yangtze River Delta, Shanghai, China
| | - Yang Gao
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China
| | - Jiamin Hu
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China
| | - Rongkui Luo
- Department of Pathology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China
| | - Yinglong Guo
- Department of Radiology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China
| | - Qiang Xie
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China
| | - Chaolong Yan
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China
| | - Yifan Tang
- Department of Neurosurgery, Shanghai Geriatric Medical Center, 2560 Chunsheng Road, Shanghai, China
| | - Pin Chen
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China
| | - Zijiang Yang
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China
| | - Qinqin Yu
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, P. R. China
| | - Fan Hu
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China
| | - Xiaobiao Zhang
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China.
- Department of Neurosurgery, Shanghai Geriatric Medical Center, 2560 Chunsheng Road, Shanghai, China.
- Cancer Center, Shanghai Zhongshan Hospital, Fudan University, Shanghai, China.
- The innovation and translation alliance of neuroendoscopy in the Yangtze River Delta, Shanghai, China.
- Digital Medical Research Center, Fudan University, 138 Yixueyuan Road, Shanghai, China.
| |
Collapse
|
40
|
Ben Rejeb S, Aloui D, Ayari A, Chouchen A. Prognostic Significance of C-MYC and EGFR Overexpression in Gastrointestinal Stromal Tumors: An Immunohistochemical Study. Appl Immunohistochem Mol Morphol 2025; 33:43-48. [PMID: 39636317 DOI: 10.1097/pai.0000000000001235] [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: 11/30/2023] [Accepted: 10/14/2024] [Indexed: 12/07/2024]
Abstract
INTRODUCTION In addition to mutations in KIT and PDGFRA, many other genetic alterations have been described in gastrointestinal stromal tumors (GISTs), including amplifications of C-MYC and EGFR, which are often associated with increased protein expression. The main of this study was to investigate the prognostic significance of C-MYC and EGFR expression in GISTs using immunohistochemistry (IHC). METHODS We collected all GIST cases over a 16-year period. These cases were tested using antibodies against C-MYC (Leica, clone EP121) and EGFR (Leica, clone 113). C-MYC staining was assessed using the H-score method for nuclear, cytoplasmic, and combined staining. For EGFR staining (either cytoplasmic or nuclear), the intensity was graded as follows: 0 (no staining), 1 (weak staining), 2 (moderate staining), and 3 (strong staining). The percentage of positive cells was evaluated using a semiquantitative approach. Statistical analysis was performed using SPSS24. RESULTS A total of 37 cases were included in our study. Nuclear expression of C-MYC was observed in 43% of the cases, with a high H-score in 43%. A statistically significant association was found between a high nuclear H-score for C-MYC and mitotic rate (P=0.046), as well as a high Ki-67 proliferation rate (P=0.046). However, no statistically significant associations were identified between the nuclear H-score of C-MYC and other clinical, pathologic, or survival data. Cytoplasmic expression of C-MYC was noted in 22% of cases, but no significant correlations were found with the clinicopathological data. EGFR staining was observed in 86% of cases, with a high score of 51%. EGFR expression was significantly associated with the mitotic index (P=0.012) and Ki-67 proliferation rate (P=0.046). CONCLUSIONS Our findings suggest that both C-MYC and EGFR may be overexpressed and/or amplified in GISTs, indicating their potential prognostic role. This could also pave the way for therapeutic strategies targeting these proteins.
Collapse
Affiliation(s)
| | | | - Asma Ayari
- Department of Pathology, Rabta's Hospital, Marsa, Tunisia
| | | |
Collapse
|
41
|
Wang C, Ma X. The role of acetylation and deacetylation in cancer metabolism. Clin Transl Med 2025; 15:e70145. [PMID: 39778006 PMCID: PMC11706801 DOI: 10.1002/ctm2.70145] [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: 08/19/2024] [Revised: 12/02/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
As a hallmark of cancer, metabolic reprogramming adjusts macromolecular synthesis, energy metabolism and redox homeostasis processes to adapt to and promote the complex biological processes of abnormal growth and proliferation. The complexity of metabolic reprogramming lies in its precise regulation by multiple levels and factors, including the interplay of multiple signalling pathways, precise regulation of transcription factors and dynamic adjustments in metabolic enzyme activity. In this complex regulatory network, acetylation and deacetylation, which are important post-translational modifications, regulate key molecules and processes related to metabolic reprogramming by affecting protein function and stability. Dysregulation of acetylation and deacetylation may alter cancer cell metabolic patterns by affecting signalling pathways, transcription factors and metabolic enzyme activity related to metabolic reprogramming, increasing the susceptibility to rapid proliferation and survival. In this review, we focus on discussing how acetylation and deacetylation regulate cancer metabolism, thereby highlighting the central role of these post-translational modifications in metabolic reprogramming, and hoping to provide strong support for the development of novel cancer treatment strategies. KEY POINTS: Protein acetylation and deacetylation are key regulators of metabolic reprogramming in tumour cells. These modifications influence signalling pathways critical for tumour metabolism. They modulate the activity of transcription factors that drive gene expression changes. Metabolic enzymes are also affected, altering cellular metabolism to support tumour growth.
Collapse
Affiliation(s)
- Cuicui Wang
- Department of Obstetrics and GynecologyShengjing Hospital of China Medical UniversityShenyang CityLiaoning ProvinceChina
- Key Laboratory of Gynecological Oncology of Liaoning ProvinceDepartment of Obstetrics and GynecologyShengjing Hospital of China Medical UniversityShenyangLiaoning ProvinceChina
| | - Xiaoxin Ma
- Department of Obstetrics and GynecologyShengjing Hospital of China Medical UniversityShenyang CityLiaoning ProvinceChina
- Key Laboratory of Gynecological Oncology of Liaoning ProvinceDepartment of Obstetrics and GynecologyShengjing Hospital of China Medical UniversityShenyangLiaoning ProvinceChina
| |
Collapse
|
42
|
Wu Y, Zhai Y, Ding Z, Xie T, Zhu W, Zhang C, Lu Y, Chen Y, Ren S, Hu Y, Li X, Zhong F, Liang Y, Wang S. Single-cell transcriptomics reveals tumor microenvironment changes and prognostic gene signatures in hepatocellular carcinoma. Int Immunopharmacol 2024; 143:113317. [PMID: 39447409 DOI: 10.1016/j.intimp.2024.113317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/23/2024] [Accepted: 10/01/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND Hepatocellular Carcinoma (HCC) is the most common type of primary liver cancer, accounting for the majority of liver cancer cases. Hepatocellular Carcinoma not only exhibits high heterogeneity but also possesses an immune-suppressive tumor microenvironment that promotes tumor evasion, posing substantial difficulties for efficient therapy. Our aim is to utilize single-cell RNA transcriptome data to investigate the dynamic changes in the tumor microenvironment during the malignant progression of HCC, the communication among immune cells, and the marker genes associated with patient prognosis. METHODS We constructed expression matrices from open single-cell RNA transcriptome data (GSE149614) of HCC patients (representing stages I-IV), establishing single-cell RNA transcriptional atlases for different stages of HCC progression. For each stage, we conducted cell subgroup analysis to identify cell types at each stage. Horizontally, we explored the dynamic changes of the same cell type across different stages, performing trajectory analysis and prognosis analysis. Vertically, we investigated pairwise comparisons of different stages of HCC progression, probing the dynamic alterations in tumor microenvironment immune cell signaling pathways. Finally, potential drugs for the treatment of HCC were predicted based on relevant genes. FINDINGS As the HCC advances towards increased malignancy, there is a shift in the predominant composition of the tumor microenvironment, with a decline in the dominance of hepatic cells. Tumor-infiltrating immune cells migrate and accumulate within the tumor microenvironment, where T cells and myeloid cells display distinct patterns of change. Genes associated with cancer-associated fibroblasts (CAFs) and T cells are correlated with adverse patient outcomes. In the late stages of HCC, the tumor microenvironment is infiltrated by more myeloid-derived suppressor cells (MDSCs), and a prognostic model constructed based on genes related to myeloid cells can predict patient outcomes. Additionally, in the analysis of transcription factors, YY1 and MYC are found to be highly expressed. Cell communication analysis among tumor-infiltrating immune cells reveals significant differences in the main signaling pathways at different stages of HCC progression. Finally, drug sensitivity analysis based on key genes identifies Acetalax, Allopurinol, and Amonafide as potential candidates for HCC treatment.
Collapse
Affiliation(s)
- Yilin Wu
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China
| | - Yangyang Zhai
- Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai Research Center of Biliary Tract Disease, Department of General Surgery, Xinhua Hospital, Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhilong Ding
- Department of Hepatobiliary Surgery, The Affiliated Huaian Hospital of Xuzhou Medical University and Huai'an Second People's Hospital, Huai'an, Jiangsu, China
| | - Tong Xie
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China
| | - WeiJie Zhu
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China
| | - Cui Zhang
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China
| | - Ying Lu
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China
| | - Yunli Chen
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China
| | - Shiying Ren
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China
| | - Yihuai Hu
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China
| | - Xiangqian Li
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China
| | - Fei Zhong
- Department of Laboratory Medicine, The Affiliated Huaian Hospital of Xuzhou Medical University and Huai'an Second People's Hospital, Huai'an, Jiangsu, China.
| | - Yong Liang
- Department of Laboratory Medicine, The Affiliated Huaian Hospital of Xuzhou Medical University and Huai'an Second People's Hospital, Huai'an, Jiangsu, China.
| | - Shiyan Wang
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China.
| |
Collapse
|
43
|
Oliveira JC, Negreiro JM, Nunes FM, Barbosa FG, Mafezoli J, Mattos MC, Fernandes MCR, Pessoa C, Furtado CLM, Zanatta G, Oliveira MCF. In Silico Study of the Anti-MYC Potential of Lanostane-Type Triterpenes. ACS OMEGA 2024; 9:50844-50858. [PMID: 39741863 PMCID: PMC11683602 DOI: 10.1021/acsomega.4c10201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 11/30/2024] [Accepted: 12/05/2024] [Indexed: 01/03/2025]
Abstract
One of the most investigated molecular targets for anticancer therapy is the proto-oncogene MYC, which is amplified and thus overexpressed in many types of cancer. Due to its structural characteristics, developing inhibitors for the target has proven to be challenging. In this study, the anti-MYC potential of lanostane-type triterpenes was investigated for the first time, using computational approaches that involved ensemble docking, prediction of structural properties and pharmacokinetic parameters, molecular dynamics (MD), and binding energy calculation using the molecular mechanics-generalized born surface area (MM-GBSA) method. The analysis of physicochemical properties, druglikeness, and pharmacokinetic parameters showed that ligands ganoderic acid E (I), ganoderlactone D (II), ganoderic acid Y (III), ganoderic acid Df (IV), lucidenic acid F (V), ganoderic acid XL4 (VI), mariesiic acid A (VII), and phellinol E (VIII) presented properties within the filter used. These eight ligands, in general, could interact with the molecular target favorably, with interaction energy values between -8.3 and -8.6 kcal mol-1. In MD, the results of RMSD, RMSF, radius of gyration, and hydrogen bonds of the complexes revealed that ligands I, IV, VI, and VII interacted satisfactorily with the protein during the simulations and assisted in its conformational and energetic stabilization. The binding energy calculation using the MM-GBSA method showed better results for the MYC-VII and MYC-I complexes (-44.98 and -41.96 kcal mol-1, respectively). These results support the hypothesis that such molecules can interact with MYC for a considerable period, which would be an essential condition for them to exert their inhibitory activity effectively.
Collapse
Affiliation(s)
- José
A. C. Oliveira
- Department
of Organic and Inorganic Chemistry, Science Center, Federal University of Ceará, Fortaleza, CE 60455-760, Brazil
| | - Jonatas M. Negreiro
- Department
of Organic and Inorganic Chemistry, Science Center, Federal University of Ceará, Fortaleza, CE 60455-760, Brazil
| | - Fátima M. Nunes
- Department
of Organic and Inorganic Chemistry, Science Center, Federal University of Ceará, Fortaleza, CE 60455-760, Brazil
| | - Francisco G. Barbosa
- Department
of Organic and Inorganic Chemistry, Science Center, Federal University of Ceará, Fortaleza, CE 60455-760, Brazil
| | - Jair Mafezoli
- Department
of Organic and Inorganic Chemistry, Science Center, Federal University of Ceará, Fortaleza, CE 60455-760, Brazil
| | - Marcos C. Mattos
- Department
of Organic and Inorganic Chemistry, Science Center, Federal University of Ceará, Fortaleza, CE 60455-760, Brazil
| | - Maria C. R. Fernandes
- Drug Research
and Development Center, Federal University
of Ceará, Rua Coronel Nunes de Melo, 1000, Fortaleza, CE 60430-275, Brazil
| | - Claudia Pessoa
- Drug Research
and Development Center, Federal University
of Ceará, Rua Coronel Nunes de Melo, 1000, Fortaleza, CE 60430-275, Brazil
| | - Cristiana L. M. Furtado
- Drug Research
and Development Center, Federal University
of Ceará, Rua Coronel Nunes de Melo, 1000, Fortaleza, CE 60430-275, Brazil
- Graduate
Program in Medical Sciences, University
of Fortaleza, Rua Francisco
Segundo da Costa, 23-57, Fortaleza, CE 60811-650, Brazil
| | - Geancarlo Zanatta
- Department
of Biophysics, Bioscience Institute, Federal
University of Rio Grande do Sul, Av. Bento Gonçalves, 9500, Building 43422, Laboratory
204, Porto Alegre, RS 91501-970, Brazil
| | - Maria C. F. Oliveira
- Department
of Organic and Inorganic Chemistry, Science Center, Federal University of Ceará, Fortaleza, CE 60455-760, Brazil
| |
Collapse
|
44
|
Siokatas C, Lampropoulou A, Smina A, Soupsana K, Kontostathi M, Karra AV, Karampelas T, Politou AS, Christoforidis S, Tamvakopoulos C, Sarli V. Developing MYC Degraders Bearing the Von Hippel-Lindau Ligand to Target the "Undruggable" MYC. ACS Pharmacol Transl Sci 2024; 7:3955-3968. [PMID: 39698270 PMCID: PMC11650737 DOI: 10.1021/acsptsci.4c00452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/15/2024] [Accepted: 11/04/2024] [Indexed: 12/20/2024]
Abstract
Although small-molecule inhibitors with moderate efficacy targeting MYC have been previously described, to this point, research efforts have failed to bring a suitable small-molecule MYC inhibitor to the clinic. Herein, the discovery of a series of novel MYC degraders bearing VHL to target the "undruggable" MYC is presented. The molecules are based on connecting a known MYC binder to a VHL ligand or pomalidomide to induce MYC degradation in various cancer cells known to express MYC. Representative compounds from our work induced MYC degradation in a time- and dose-dependent manner. Selected compounds, CSI86 and CSI107, displayed antiproliferative activity (IC50 values of 13-18 μM) against breast and prostate cancer cells. The lead molecules were further evaluated in terms of cell uptake, potential to degrade MYC, and pharmacokinetics in mice. Encouraging results presented herein suggest that the presented analogs may serve as prototype structures of future therapeutic agents for the treatment of MYC-dependent tumors. MYC protein degraders can well complement the more established inhibition approaches that have been presented in the past (e.g., disruption of the MYC-MAX complex formation by small-molecule inhibitors).
Collapse
Affiliation(s)
- Christos Siokatas
- Vasiliki
Sarli - Department of Chemistry, Aristotle
University of Thessaloniki, University Campus, Thessaloniki 54124, Greece
| | - Alexandra Lampropoulou
- Constantin
Tamvakopoulos - Center of Clinical Research, Experimental Surgery
and Translational Research, Division of Pharmacology-Pharmacotechnology, Biomedical Research Foundation, Academy of Athens, Soranou Ephessiou Street 4, Athens 11527, Greece
| | - Alexandra Smina
- Constantin
Tamvakopoulos - Center of Clinical Research, Experimental Surgery
and Translational Research, Division of Pharmacology-Pharmacotechnology, Biomedical Research Foundation, Academy of Athens, Soranou Ephessiou Street 4, Athens 11527, Greece
| | - Katerina Soupsana
- Laboratory
of Biological Chemistry, Department of Medicine, School of Health Sciences, University of Ioannina, Ioannina 45110, Greece
| | - Martha Kontostathi
- Laboratory
of Biological Chemistry, Department of Medicine, School of Health Sciences, University of Ioannina, Ioannina 45110, Greece
- Biomedical
Research Institute, Foundation for Research
and Technology, Ioannina 45110, Greece
| | - Athina-Vasiliki Karra
- Laboratory
of Biological Chemistry, Department of Medicine, School of Health Sciences, University of Ioannina, Ioannina 45110, Greece
- Biomedical
Research Institute, Foundation for Research
and Technology, Ioannina 45110, Greece
| | - Theodoros Karampelas
- Constantin
Tamvakopoulos - Center of Clinical Research, Experimental Surgery
and Translational Research, Division of Pharmacology-Pharmacotechnology, Biomedical Research Foundation, Academy of Athens, Soranou Ephessiou Street 4, Athens 11527, Greece
| | - Anastasia S. Politou
- Laboratory
of Biological Chemistry, Department of Medicine, School of Health Sciences, University of Ioannina, Ioannina 45110, Greece
- Biomedical
Research Institute, Foundation for Research
and Technology, Ioannina 45110, Greece
| | - Savvas Christoforidis
- Laboratory
of Biological Chemistry, Department of Medicine, School of Health Sciences, University of Ioannina, Ioannina 45110, Greece
- Biomedical
Research Institute, Foundation for Research
and Technology, Ioannina 45110, Greece
| | - Constantin Tamvakopoulos
- Constantin
Tamvakopoulos - Center of Clinical Research, Experimental Surgery
and Translational Research, Division of Pharmacology-Pharmacotechnology, Biomedical Research Foundation, Academy of Athens, Soranou Ephessiou Street 4, Athens 11527, Greece
| | - Vasiliki Sarli
- Vasiliki
Sarli - Department of Chemistry, Aristotle
University of Thessaloniki, University Campus, Thessaloniki 54124, Greece
| |
Collapse
|
45
|
Shnaider PV, Malyants IK, Ivanova OM, Gordeeva VS, Svirina EA, Zakharzhevskaya NB, Shagaleeva OY, Selezneva OV, Bogomazova AN, Lukina MM, Aleshikova OI, Babaeva NA, Slonov AV, Shender VO. Establishment of Novel High-Grade Serous Ovarian Carcinoma Cell Line OVAR79. Int J Mol Sci 2024; 25:13236. [PMID: 39769003 PMCID: PMC11676670 DOI: 10.3390/ijms252413236] [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: 11/16/2024] [Revised: 12/03/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
High-grade serous ovarian carcinoma (HGSOC) remains the most common and deadly form of ovarian cancer. However, available cell lines usually fail to appropriately represent its complex molecular and histological features. To overcome this drawback, we established OVAR79, a new cell line derived from the ascitic fluid of a patient with a diagnosis of HGSOC, which adds a unique set of properties to the study of ovarian cancer. In contrast to the common models, OVAR79 expresses TP53 without the common hotspot mutations and harbors the rare combination of mutations in both PIK3CA and PTEN genes, together with high-grade chromosomal instability with multiple gains and losses. These features, together with the high proliferation rate, ease of cultivation, and exceptional transfection efficiency of OVAR79, make it a readily available and versatile tool for various studies in the laboratory. We extensively characterized its growth, migration, and sensitivity to platinum- and taxane-based treatments in comparison with the commonly used SKOV3 and OVCAR3 ovarian cell lines. In summary, OVAR79 is an excellent addition for basic and translational ovarian cancer research and offers new insights into the biology of HGSOC.
Collapse
Affiliation(s)
- Polina V. Shnaider
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435 Moscow, Russia
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, 119435 Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Irina K. Malyants
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, 119435 Moscow, Russia
| | - Olga M. Ivanova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435 Moscow, Russia
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, 119435 Moscow, Russia
- Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
| | - Veronika S. Gordeeva
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435 Moscow, Russia
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, 119435 Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia
| | - Ekaterina A. Svirina
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, 119435 Moscow, Russia
| | - Natalya B. Zakharzhevskaya
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, 119435 Moscow, Russia
| | - Olga Y. Shagaleeva
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, 119435 Moscow, Russia
| | - Oksana V. Selezneva
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, 119435 Moscow, Russia
| | - Alexandra N. Bogomazova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435 Moscow, Russia
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, 119435 Moscow, Russia
| | - Maria M. Lukina
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435 Moscow, Russia
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, 119435 Moscow, Russia
| | - Olga I. Aleshikova
- National Medical Scientific Centre of Obstetrics, Gynaecology and Perinatal Medicine Named After V.I. Kulakov, 117198 Moscow, Russia
| | - Nataliya A. Babaeva
- National Medical Scientific Centre of Obstetrics, Gynaecology and Perinatal Medicine Named After V.I. Kulakov, 117198 Moscow, Russia
| | - Andrey V. Slonov
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, 119435 Moscow, Russia
| | - Victoria O. Shender
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435 Moscow, Russia
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, 119435 Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia
| |
Collapse
|
46
|
Endicott SJ. Chaperone-mediated autophagy as a modulator of aging and longevity. FRONTIERS IN AGING 2024; 5:1509400. [PMID: 39687864 PMCID: PMC11647017 DOI: 10.3389/fragi.2024.1509400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024]
Abstract
Chaperone-mediated autophagy (CMA) is the lysosomal degradation of individually selected proteins, independent of vesicle fusion. CMA is a central part of the proteostasis network in vertebrate cells. However, CMA is also a negative regulator of anabolism, and it degrades enzymes required for glycolysis, de novo lipogenesis, and translation at the cytoplasmic ribosome. Recently, CMA has gained attention as a possible modulator of rodent aging. Two mechanistic models have been proposed to explain the relationship between CMA and aging in mice. Both of these models are backed by experimental data, and they are not mutually exclusionary. Model 1, the "Longevity Model," states that lifespan-extending interventions that decrease signaling through the INS/IGF1 signaling axis also increase CMA, which degrades (and thereby reduces the abundance of) several proteins that negatively regulate vertebrate lifespan, such as MYC, NLRP3, ACLY, and ACSS2. Therefore, enhanced CMA, in early and midlife, is hypothesized to slow the aging process. Model 2, the "Aging Model," states that changes in lysosomal membrane dynamics with age lead to age-related losses in the essential CMA component LAMP2A, which in turn reduces CMA, contributes to age-related proteostasis collapse, and leads to overaccumulation of proteins that contribute to age-related diseases, such as Alzheimer's disease, Parkinson's disease, cancer, atherosclerosis, and sterile inflammation. The objective of this review paper is to comprehensively describe the data in support of both of these explanatory models, and to discuss the strengths and limitations of each.
Collapse
Affiliation(s)
- S. Joseph Endicott
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
- Autophagy, Inflammation, and Metabolism Center of Biomedical Research Excellence, (AIM CoBRE), University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| |
Collapse
|
47
|
Hu F, Zhao L, Wang J, Li X, Xue Z, Ma Y, Zheng M, Chen C, Tong M, Guo X, Li H, Jin H, Xie Q, Zhang X, Huang C, Huang H. TRIM40 interacts with ROCK1 directly and inhibits colorectal cancer cell proliferation through the c-Myc/p21 axis. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119855. [PMID: 39357549 DOI: 10.1016/j.bbamcr.2024.119855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 09/10/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Colorectal cancer (CRC) is the most common malignancy of the digestive tract, and to date, morbidity and mortality rates remain high. While existing therapeutic methods have achieved certain effective outcomes, there are still many problems in treating this disease. Therefore, it is still urgent to constantly find new therapeutic targets in CRC that could lead to new therapeutics. METHODS Immunohistochemistry, Real-time PCR and Western Blot were employed to measure mRNA and protein levels of the target protein, respectively. The proliferation ability of CRC cells was evaluated using ATP assay, Soft agar assay, and nude mouse subcutaneous tumorigenesis assay. Protein Degradation Assay was conducted to determine protein degradation rate, while Ubiquitination assay was used to assess the ubiquitination modification level of target proteins. Immunoprecipitation assay was used to study protein interactions, and pull-down assay was employed to investigate direct interactions between proteins. RESULTS TRIM40 was significantly down-regulated in CRC tissues, with its expression levels positively correlating with disease prognosis. Using both in vitro and in vivo approaches, it was demonstrated that TRIM40 could significantly inhibit the proliferation of CRC cells. Molecular mechanism studies showed that TRIM40 directly binds to and ubiquitinates ROCK1 protein, accelerating its degradation and subsequently reducing the stability of c-Myc protein. This cascade of events results in the release of transcriptional inhibition of p21 by c-Myc, leading to increased p21 expression and G0/G1 phase arrest in CRC cells. CONCLUSION This research suggests that TRIM40 could be a valuable therapeutic target for the treatment of CRC.
Collapse
Affiliation(s)
- Fangyu Hu
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Lingling Zhao
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Junyu Wang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xiaoying Li
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Zixuan Xue
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yimeng Ma
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Minghui Zheng
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Chenglin Chen
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Meiting Tong
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xiaohuan Guo
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Hongyan Li
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Honglei Jin
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Qipeng Xie
- Department of Laboratory Medicine, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xiaodong Zhang
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Chuanshu Huang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325035, China.
| | - Haishan Huang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| |
Collapse
|
48
|
Masciale V, Banchelli F, Grisendi G, Samarelli AV, Raineri G, Rossi T, Zanoni M, Cortesi M, Bandini S, Ulivi P, Martinelli G, Stella F, Dominici M, Aramini B. The molecular features of lung cancer stem cells in dedifferentiation process-driven epigenetic alterations. J Biol Chem 2024; 300:107994. [PMID: 39547513 PMCID: PMC11714729 DOI: 10.1016/j.jbc.2024.107994] [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: 03/23/2024] [Revised: 10/30/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024] Open
Abstract
Cancer stem cells (CSCs) may be dedifferentiated somatic cells following oncogenic processes, representing a subpopulation of cells able to promote tumor growth with their capacities for proliferation and self-renewal, inducing lineage heterogeneity, which may be a main cause of resistance to therapies. It has been shown that the "less differentiated process" may have an impact on tumor plasticity, particularly when non-CSCs may dedifferentiate and become CSC-like. Bidirectional interconversion between CSCs and non-CSCs has been reported in other solid tumors, where the inflammatory stroma promotes cell reprogramming by enhancing Wnt signaling through nuclear factor kappa B activation in association with intracellular signaling, which may induce cells' pluripotency, the oncogenic transformation can be considered another important aspect in the acquisition of "new" development programs with oncogenic features. During cell reprogramming, mutations represent an initial step toward dedifferentiation, in which tumor cells switch from a partially or terminally differentiated stage to a less differentiated stage that is mainly manifested by re-entry into the cell cycle, acquisition of a stem cell-like phenotype, and expression of stem cell markers. This phenomenon typically shows up as a change in the form, function, and pattern of gene and protein expression, and more specifically, in CSCs. This review would highlight the main epigenetic alterations, major signaling pathways and driver mutations in which CSCs, in tumors and specifically, in lung cancer, could be involved, acting as key elements in the differentiation/dedifferentiation process. This would highlight the main molecular mechanisms which need to be considered for more tailored therapies.
Collapse
Affiliation(s)
- Valentina Masciale
- Laboratory of Cellular Therapies, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
| | - Federico Banchelli
- Department of Statistical Sciences "Paolo Fortunati", Alma Mater Studiorum- University of Bologna, Bologna, Italy
| | - Giulia Grisendi
- Laboratory of Cellular Therapies, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
| | - Anna Valeria Samarelli
- Laboratory of and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
| | - Giulia Raineri
- Laboratory of Cellular Therapies, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
| | - Tania Rossi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Michele Zanoni
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Michela Cortesi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Sara Bandini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Paola Ulivi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Giovanni Martinelli
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Franco Stella
- Thoracic Surgery Unit, Department of Medical and Surgical Sciences-DIMEC of the Alma Mater Studiorum, University of Bologna, G.B. Morgagni-L. Pierantoni Hospital, Forlì, Italy
| | - Massimo Dominici
- Laboratory of Cellular Therapies, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy; Division of Oncology, University Hospital of Modena and Reggio Emilia, University of Modena and Reggio Emilia, Modena, Italy
| | - Beatrice Aramini
- Thoracic Surgery Unit, Department of Medical and Surgical Sciences-DIMEC of the Alma Mater Studiorum, University of Bologna, G.B. Morgagni-L. Pierantoni Hospital, Forlì, Italy.
| |
Collapse
|
49
|
Du B, Chen M, Chang L, Zhang X, Zhang X, Wang X, Gong P, Zhang N, Zhang X, Li X, Li J. Immunization with the NcMYR1 gene knockout strain effectively protected C57BL/6 mice and their pups against the Neospora caninum challenge. Virulence 2024; 15:2427844. [PMID: 39607301 PMCID: PMC11610562 DOI: 10.1080/21505594.2024.2427844] [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: 02/25/2024] [Revised: 10/16/2024] [Accepted: 10/31/2024] [Indexed: 11/29/2024] Open
Abstract
Neospora caninum is an important protozoan parasite that causes abortion in cattle and nervous system dysfunction in dogs. No effective drugs and vaccines for neosporosis are available. Further elucidation of proteins related to N. caninum virulence will provide potential candidates for vaccine development against neosporosis. In the present study, N. caninum c-Myc regulatory protein (NcMYR1) gene knockout strains (ΔNcMYR1-1, ΔNcMYR1-2, and ΔNcMYR1-3) were generated using the CRISPR-Cas9 gene editing system to investigate phenotype changes and the potential of the ΔNcMYR1-1 strain as an attenuated vaccine, and this is the first time of using the N. caninum CRISPR-Cas9 gene knockout strain as an attenuated vaccine. NcMYR1 was determined to be a cytoplasmic protein in N. caninum tachyzoites. The deficiency of NcMYR1 decreased the plaque area and the rate of invasion, replication, and egression of the parasites. ΔNcMYR1-1 strain-infected C57BL/6 mice had 100% survival rate, reduced parasite burden, and alleviated pathological changes in tissues compared with those in Nc-1 strain-infected mice. Immunization with ΔNcMYR1-1 tachyzoites increased the productions of cytokines in mice, with a survival rate reaching 80%, and the parasite burdens in the liver and spleen were greatly reduced when challenged with the Nc-1 strain with a lethal dose after 40 days of ΔNcMYR1-1 tachyzoite immunization. ΔNcMYR1 immunization could decrease the abortion rate of female mice from 71.4% to 12.5% and increase the survival rate of pups from 12.5% to 83.3% against the N. caninum challenge. Above all, NcMYR1 is a virulence factor and the ΔNcMYR1-1 strain could be used as a candidate vaccine against N. caninum infection and vertical transmission.
Collapse
Affiliation(s)
- Boya Du
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Mengge Chen
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Le Chang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xu Zhang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xuancheng Zhang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xiaocen Wang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Pengtao Gong
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Nan Zhang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xichen Zhang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xin Li
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Jianhua Li
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
50
|
Puxeddu M, Ling L, Ripa S, D'Ambrosio M, Nalli M, Parisi A, Sciò P, Coluccia A, Granese A, Santelli M, Masci D, Cuřínová P, Naro C, Sette C, Pastore A, Stornaiuolo M, Bigogno C, Dondio G, Di Magno L, Canettieri G, Liu T, Silvestri R, La Regina G. Development of N-(4-(1 H-Imidazol-1-yl)phenyl)-4-chlorobenzenesulfonamide, a Novel Potent Inhibitor of β-Catenin with Enhanced Antitumor Activity and Metabolic Stability. J Med Chem 2024; 67:20298-20314. [PMID: 39508273 DOI: 10.1021/acs.jmedchem.4c01708] [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: 11/15/2024]
Abstract
The potential as a cancer therapeutic target of the recently reported hotspot binding region close to Lys508 of the β-catenin armadillo repeat domain was not exhaustively explored. In order to get more insight, we synthesized novel N-(heterocyclylphenyl)benzenesulfonamides 6-28. The new compounds significantly inhibited Wnt-dependent transcription as well as SW480 and HCT116 cancer cell proliferation. Compound 25 showed binding mode consistent with this hotspot binding region. Compound 25 inhibited the growth of SW480 and HCT116 cancer cells with IC50's of 2 and 0.12 μM, respectively, and was superior to the reference compounds 5 and 5-FU. 25 inhibited the growth of HCT-116 xenografted in BALB/Cnu/nu mice, reduced the expression of the proliferation marker Ki67, and significantly affected the expression of cancer-related genes. After incubation with human and mouse liver microsomes, 25 showed a higher metabolic stability than 5. Compound 25 aims to be a promising lead for the development of colorectal cancer anticancer therapies.
Collapse
Affiliation(s)
- Michela Puxeddu
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| | - Lele Ling
- Department of Acupuncture and Moxibustion, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, 200086 Shanghai, China
| | - Silvia Ripa
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy
| | - Michele D'Ambrosio
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| | - Marianna Nalli
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| | - Anastasia Parisi
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| | - Pietro Sciò
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| | - Antonio Coluccia
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| | - Arianna Granese
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| | - Martina Santelli
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Catholic University of the Sacred Heart, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Domiziana Masci
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Catholic University of the Sacred Heart, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Petra Cuřínová
- Department of Organic Chemistry, University of Chemistry and Technology Prague, Technická 5, 16628 Prague 6, Czech Republic
| | - Chiara Naro
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Catholic University of the Sacred Heart, Largo Francesco Vito 1, 00168 Rome, Italy
- GSTeP-Organoids Research Core Facility, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Claudio Sette
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Catholic University of the Sacred Heart, Largo Francesco Vito 1, 00168 Rome, Italy
- GSTeP-Organoids Research Core Facility, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Arianna Pastore
- Department of Pharmacy, University of Naples "Federico II", Via Domenico Montesano 49, 80131 Naples, Italy
| | - Mariano Stornaiuolo
- Department of Pharmacy, University of Naples "Federico II", Via Domenico Montesano 49, 80131 Naples, Italy
| | - Chiara Bigogno
- Aphad SrL, Via della Resistenza 65, 20090 Buccinasco, Italy
| | - Giulio Dondio
- Aphad SrL, Via della Resistenza 65, 20090 Buccinasco, Italy
| | - Laura Di Magno
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy
| | - Gianluca Canettieri
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy
| | - Te Liu
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 365 South Xiangyang Road, 200031 Shanghai, China
| | - Romano Silvestri
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| | - Giuseppe La Regina
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| |
Collapse
|