1
|
Xu L, Shi M, Qin G, Lin X, Huang B. Environmental pollutant Di-(2-ethylhexyl) phthalate induces asthenozoospermia: new insights from network toxicology. Mol Divers 2025; 29:2179-2192. [PMID: 39259422 DOI: 10.1007/s11030-024-10976-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 08/20/2024] [Indexed: 09/13/2024]
Abstract
The global decline in sperm quality in men is closely associated with environmental exposure to the plasticizer Di-(2-ethylhexyl) phthalate (DEHP), but the molecular mechanisms underlying its induction of asthenozoospermia (AZS) remain incompletely understood. By integrating the toxicological targets of DEHP and differential genes in AZS patients, and combining machine learning, molecular docking, and dynamics simulations, this study successfully identified hub genes and signaling pathways induced by DEHP in AZS, aiming to provide new strategies for the prevention and treatment of this disease. A total of 26 toxicological targets were identified, with FGFR1, MMP7, and ST14 clearly defined as playing crucial regulatory roles in DEHP-induced AZS. This study also reveals that DEHP may induce reproductive system inflammation, affecting the proliferation and survival of reproductive cells, and subsequently impacting sperm vitality, possibly through regulating the mTORC1 pathway, TNF-α signaling via the NF-κB pathway, and MYC targets v1 pathway. Furthermore, changes in the immune microenvironment revealed the significant impact of immune status on testicular function. In conclusion, this study provides important scientific evidence for understanding the molecular mechanisms of AZS and developing prevention and treatment strategies based on toxicological targets.
Collapse
Affiliation(s)
- Lei Xu
- The First School of Clinical Medicine, College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, Yunnan, China
| | - Menghua Shi
- The First School of Clinical Medicine, College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, Yunnan, China
| | - Guozheng Qin
- The First School of Clinical Medicine, College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, Yunnan, China
- Yunnan Provincial Hospital of Chinese Medicine, Kunming, 650021, Yunnan, China
| | - Xuyao Lin
- The First School of Clinical Medicine, College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, Yunnan, China.
| | - Bin Huang
- The First School of Clinical Medicine, College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, Yunnan, China.
- School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua, 418000, Hunan, China.
| |
Collapse
|
2
|
Sun Y, Li Y, Zhang A, Hu T, Li M. Prognostic model identification of ribosome biogenesis-related genes in pancreatic cancer based on multiple machine learning analyses. Discov Oncol 2025; 16:905. [PMID: 40411705 PMCID: PMC12103412 DOI: 10.1007/s12672-025-02733-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 05/16/2025] [Indexed: 05/26/2025] Open
Abstract
BACKGROUND Pancreatic cancer is a highly aggressive cancer characterized by low survival rate. Enhanced ribosome biogenesis may be associated with tumor drug resistance and malignant phenotypes, representing a potential therapeutic target in pancreatic cancer. Therefore, exploring the molecular mechanisms of ribosome biogenesis in pancreatic cancer may uncover new biomarkers and potential therapeutic targets, facilitating the development of personalized treatment strategies. METHODS Ribosome biogenesis-related gene signatures were acquired from TCGA and Gene Cards databases. Prognostic gene sets were screened using machine learning algorithms to construct a risk model, which was externally validated via GEO database. Single-cell RNA sequencing analysis (GSE155698 dataset) was performed to assess gene expression patterns and module scores. RESULTS Sixty ribosome biogenesis-related prognostic genes were identified in pancreatic cancer. Cox regression and machine learning algorithms selected nine pivotal biomarkers (ECT2; CKB; HMGA2; TPX2; ERBB3; SLC2A1; KRT13; PRSS3; CRABP2) with high diagnostic and prognostic specificity for PAAD. The machine learning-derived risk score correlated strongly with tumor proliferation pathways and immunosuppression, suggesting dual roles in tumor promotion and immunosuppressive microenvironment remodeling. Single-cell analysis highlighted predominant expression of CKB, SLC2A1, ERBB3, CRABP2, and PRSS3 in pancreatic ductal epithelial cells. CONCLUSIONS Our results shed light on the potential connections between ribosome biogenesis-related molecular characteristics and clinical features, the tumor microenvironment, and clinical drug responses. The research underscores the critical role of ribosome biogenesis in the progression and treatment resistance of pancreatic cancer, offering valuable new perspectives for prognostic evaluation and therapeutic response prediction in pancreatic cancer.
Collapse
Affiliation(s)
- Yuan Sun
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yan Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Anlan Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tao Hu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ming Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
3
|
Kuo HJ, Srinivasan P, Lin YC, Lu M, Rungkittikhun C, Zhang Q, Hu WS. Transcriptomic functional characterization of recombinant adeno-associated virus producing cell line adapted to suspension-growth. Biotechnol Prog 2025:e70042. [PMID: 40396307 DOI: 10.1002/btpr.70042] [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: 02/27/2025] [Revised: 04/23/2025] [Accepted: 05/07/2025] [Indexed: 05/22/2025]
Abstract
Recombinant adeno-associated virus (rAAV) is a widely used delivery vehicle in gene therapy. A scalable production technology is essential for its wide clinical applications. We have taken a synthetic biology approach to generate HEK293-based cell lines which harbor integrated genetic elements encoding essential AAV and adenoviral helper components and can be induced to produce rAAV. Through cycles of cell line enhancement, a high rAAV productivity could be achieved. The cell lines, like their parental HEK293, grew adherently. For scalable production, cell cultivation in suspension is highly desirable. A producer cell line GX6B was adapted to suspension growth in serum-free medium (named GX6Bs). However, it had substantially reduced virus titer. Returning GX6Bs cells to adherent culture conditions using adherent medium and cultured stationarily brought the productivity back to close to the level of adherent GX6B. A survey of the transcriptome revealed that induction and rAAV production elicited a wide range of cellular changes in various functional classes, including host immune defense response and nucleosome organization. The response was more subdued in suspension-growing GX6Bs. Upon reverting to adherent growth, the cellular transcriptome change regained its vigor to be more similar to that seen in GX6B. The GX6Bs maintained in suspension serum-free conditions were then reverted to the adherent culture medium but under an agitated culture environment to keep suspension growth for rAAV production. The productivity returned to within 25%-50% of GX6B. This work demonstrated the feasibility of the suspension culture of synthetic cell lines for the expansion and production of rAAV.
Collapse
Affiliation(s)
- Han-Jung Kuo
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota, USA
| | - Prahalad Srinivasan
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota, USA
| | - Yu-Chieh Lin
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota, USA
| | - Min Lu
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota, USA
| | - Carissa Rungkittikhun
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota, USA
| | - Qi Zhang
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota, USA
| | - Wei-Shou Hu
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
4
|
Zhang W, Wang R, Guo R, Wang Y, Wang H, Li Y, Li X, Song J. The potential of secretogranin V as a prognostic biomarker in non-small cell lung cancer. Sci Rep 2025; 15:16589. [PMID: 40360599 PMCID: PMC12075651 DOI: 10.1038/s41598-025-00747-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 04/30/2025] [Indexed: 05/15/2025] Open
Abstract
Recent studies indicate that Secretogranin V (SCG5) is aberrantly expressed in various cancers and may be linked to tumor progression and prognosis. This study aims to evaluate the potential of SCG5 as a prognostic biomarker for non-small cell lung cancer (NSCLC). We employed a combination of bioinformatics analysis, Western blotting, and immunofluorescence techniques to investigate the role of SCG5 in NSCLC. A comprehensive analysis of TCGA and GEO pan-cancer datasets revealed a consistent upregulation of SCG5 across multiple cancer types. In NSCLC, SCG5 expression was significantly higher in tumor tissues compared to normal lung tissues (p < 0.001). Kaplan-Meier survival analysis demonstrated that patients with elevated SCG5 expression exhibited lower overall survival rates, suggesting a strong association with poor prognosis. Univariate and multivariate COX regression analyses, conducted on both TCGA cases and our collected patient data, confirmed SCG5 as an independent prognostic factor for NSCLC. Furthermore, immune infiltration analysis indicated a significant correlation between SCG5 expression and various immune cell subpopulations, underscoring its potential role as a biomarker for adverse outcomes. Western blot analysis further validated the elevated levels of SCG5 in NSCLC tissues and cell lines compared to their normal counterparts. Based on our findings, we hypothesize that SCG5 may serve as a valuable biomarker for predicting the prognosis of non-small cell lung cancer, thereby guiding future research in the fields of diagnosis, progression, therapy, and prognosis of NSCLC.
Collapse
Affiliation(s)
- Weisong Zhang
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Medical School of Nantong University, Nantong, 226001, People's Republic of China
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People's Hospital, Yancheng, 224000, People's Republic of China
| | - Rui Wang
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Medical School of Nantong University, Nantong, 226001, People's Republic of China
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People's Hospital, Yancheng, 224000, People's Republic of China
| | - Rongqi Guo
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Medical School of Nantong University, Nantong, 226001, People's Republic of China
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People's Hospital, Yancheng, 224000, People's Republic of China
| | - Yihao Wang
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Medical School of Nantong University, Nantong, 226001, People's Republic of China
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People's Hospital, Yancheng, 224000, People's Republic of China
| | - Hao Wang
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Medical School of Nantong University, Nantong, 226001, People's Republic of China
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People's Hospital, Yancheng, 224000, People's Republic of China
| | - Yangyang Li
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Medical School of Nantong University, Nantong, 226001, People's Republic of China
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People's Hospital, Yancheng, 224000, People's Republic of China
| | - Xia Li
- Department of Respiratory Medicine, Affiliated Hospital 6 of Nantong University, Yancheng Third People's Hospital, No. 606 Xindu Road, Yandu District, Yancheng, 224000, People's Republic of China.
| | - Jianxiang Song
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Medical School of Nantong University, Nantong, 226001, People's Republic of China.
- Department of Thoracic Cardiothoracic Surgery, Yancheng Third People's Hospital, Affiliated Hospital 6 of Nantong University, No. 606 Xindu Road, Yandu District, Yancheng City, Yancheng, People's Republic of China.
| |
Collapse
|
5
|
Mulet-Lazaro R, Delwel R. One for All and All for One: New Insights into Enhancers Driving MYC Dysregulation. Blood Cancer Discov 2025; 6:149-152. [PMID: 40237701 PMCID: PMC12050961 DOI: 10.1158/2643-3230.bcd-25-0075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Accepted: 03/11/2025] [Indexed: 04/18/2025] Open
Abstract
The role of the proto-oncogene MYC as a driver of lymphoma has been known since the 1980s, but the mechanisms underlying its dysregulation are not completely understood. In this issue of Blood Cancer Discovery, Iyer and colleagues employ a CRISPR interference screen targeted at open chromatin regions to unveil enhancers critical for MYC overexpression in lymphoma, including a novel regulatory element in the MYC locus that controls germinal center reentry and is recurrently amplified in diffuse large B-cell lymphomas (germinal center B-cell diffuse large B-cell lymphoma). See related article by Iyer et al., p. 233.
Collapse
Affiliation(s)
- Roger Mulet-Lazaro
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - Ruud Delwel
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| |
Collapse
|
6
|
Pan X, Luo J, Zhu R, Peng J, Jin Y, Zhang L, Pei J. Transcriptomics-based identification of biomarkers associated with mast cell activation during ischemia-reperfusion injury in kidney transplantation. Transpl Immunol 2025; 90:102224. [PMID: 40147738 DOI: 10.1016/j.trim.2025.102224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 03/22/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Ischemia-reperfusion injury (IRI) in kidney transplantation can delay graft function recovery and increase the risk of rejection. Mast cell activation releases various bioactive mediators that exacerbate renal IRI. Assessing mast cell activation may be crucial for managing IRI after kidney transplantation. METHODS We analyzed the dataset GSE43974 from the Gene Expression Omnibus (GEO) to evaluate immune cell infiltration during the IRI phase of kidney transplantation using the CIBERSORT algorithm. Weighted gene co-expression network analysis (WGCNA) was performed to identify genes most strongly correlated with mast cell activation. Hub genes were identified using protein-protein interaction (PPI) network analysis and machine learning algorithms. Model accuracy for identifying hub genes was assessed using receiver operating characteristic (ROC) curve calibration. Clinical utility was evaluated through decision curve analysis (DCA). Correlation analysis was conducted to explore associations between the selected hub genes and immune cell infiltration. Additionally, a hub gene-miRNA regulatory network was constructed. RESULTS Mast cell activation exhibited the most significant variation among graft-infiltrating immune cells during IRI. WGCNA identified 115 genes closely associated with mast cell activation, from which three hub genes-JUN, MYC, and ALDH2-were selected using a PPI network and machine learning approach. A diagnostic model based on these three genes demonstrated high accuracy, as validated by the Hosmer-Lemeshow test (P = 0.980) and an area under the ROC curve (AUC) of 1. DCA indicated that these hub genes had strong clinical decision-making relevance, while correlation analysis confirmed their associations with multiple immune cell types. Finally, a hub gene-miRNA network provided a theoretical framework for the regulatory mechanisms of the three genes. CONCLUSION JUN, MYC, and ALDH2 may serve as biomarkers of mast cell activation during IRI in kidney transplantation. Further studies are warranted to explore their potential in mitigating IRI.
Collapse
Affiliation(s)
- Xingyu Pan
- Department of Nursing, Affiliated Hospital of Zunyi Medical University, Zunyi 563100, China; Nursing School of Zunyi Medical University, Zunyi 563100, China
| | - Jin Luo
- Department of Pediatric Surgery, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361005, China
| | - Rong Zhu
- Department of Pediatric surgrey, Guizhou Provincial People's Hospital, Guiyang 550002, China
| | - Jinpu Peng
- Department of Pediatric surgrey, Guizhou Provincial People's Hospital, Guiyang 550002, China
| | - Yuhan Jin
- Department of Nursing, Affiliated Hospital of Zunyi Medical University, Zunyi 563100, China; Nursing School of Zunyi Medical University, Zunyi 563100, China
| | - Li Zhang
- Department of Nursing, Affiliated Hospital of Zunyi Medical University, Zunyi 563100, China; Nursing School of Zunyi Medical University, Zunyi 563100, China.
| | - Jun Pei
- Department of Pediatric surgrey, Guizhou Provincial People's Hospital, Guiyang 550002, China.
| |
Collapse
|
7
|
Yu J, Liu D, Yuan Y, Sun C, Su Z. Rethinking MYC inhibition: a multi-dimensional approach to overcome cancer's master regulator. Front Cell Dev Biol 2025; 13:1601975. [PMID: 40365020 PMCID: PMC12069295 DOI: 10.3389/fcell.2025.1601975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Accepted: 04/21/2025] [Indexed: 05/15/2025] Open
Abstract
MYC, a master regulator in oncogenesis, has long been deemed "undruggable" due to its intrinsically disordered structure. However, recent advances are overturning this view, with direct inhibitors like Omomyc (OMO-103) and PROTAC-based degraders such as WBC100 showing promising clinical progress. Complementary strategies-including BET and CDK9 inhibitors, RNA-based therapeutics, nanobodies, and engineered proteases-are expanding the therapeutic landscape. Despite challenges in specificity, toxicity, and delivery, these innovations underscore MYC's emerging druggability. Moreover, combination therapies integrating MYC inhibitors with chemotherapy, radiotherapy, or immunotherapy demonstrate synergistic potential. This article advocates for a multi-dimensional, biomarker-guided approach to MYC targeting, emphasizing rational drug combinations and continued innovation to overcome resistance and improve outcomes in MYC-driven cancers.
Collapse
Affiliation(s)
- Jing Yu
- Qingdao Hospital, University of Health and Rehabilitation Sciences, Qingdao, Municipal Hospital, Qingdao, China
| | - Dan Liu
- Qingdao Hospital, University of Health and Rehabilitation Sciences, Qingdao, Municipal Hospital, Qingdao, China
| | - Yujian Yuan
- Qingdao Preschool Techers’ School, Qingdao, China
| | - Chunxia Sun
- Qingdao Hospital, University of Health and Rehabilitation Sciences, Qingdao, Municipal Hospital, Qingdao, China
| | - Zihan Su
- Qingdao Hospital, University of Health and Rehabilitation Sciences, Qingdao, Municipal Hospital, Qingdao, China
| |
Collapse
|
8
|
Breves SL, Di Giammartino DC, Nicholson J, Cirigliano S, Mahmood SR, Lee UJ, Martinez-Fundichely A, Jungverdorben J, Singhania R, Rajkumar S, Kirou R, Studer L, Khurana E, Polyzos A, Fine HA, Apostolou E. Three-dimensional regulatory hubs support oncogenic programs in glioblastoma. Mol Cell 2025; 85:1330-1348.e6. [PMID: 40147440 PMCID: PMC12009607 DOI: 10.1016/j.molcel.2025.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 12/18/2024] [Accepted: 03/05/2025] [Indexed: 03/29/2025]
Abstract
Dysregulation of enhancer-promoter communication in the three-dimensional (3D) nucleus is increasingly recognized as a potential driver of oncogenic programs. Here, we profiled the 3D enhancer-promoter networks of patient-derived glioblastoma stem cells to identify central regulatory nodes. We focused on hyperconnected 3D hubs and demonstrated that hub-interacting genes exhibit high and coordinated expression at the single-cell level and are associated with oncogenic programs that distinguish glioblastoma from low-grade glioma. Epigenetic silencing of a recurrent hub-with an uncharacterized role in glioblastoma-was sufficient to cause downregulation of hub-connected genes, shifts in transcriptional states, and reduced clonogenicity. Integration of datasets across 16 cancers identified "universal" and cancer-type-specific 3D hubs that enrich for oncogenic programs and factors associated with worse prognosis. Genetic alterations could explain only a small fraction of hub hyperconnectivity and increased activity. Overall, our study provides strong support for the potential central role of 3D regulatory hubs in controlling oncogenic programs and properties.
Collapse
Affiliation(s)
- Sarah L Breves
- Sanford I. Weill Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Physiology, Biophysics and Systems Biology Program, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY, USA; Department of Surgery, New York-Presbyterian Hospital, Weill Cornell Medicine, New York, NY, USA; Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Dafne Campigli Di Giammartino
- Sanford I. Weill Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - James Nicholson
- Sanford I. Weill Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Meyer Cancer Center, Division of Neuro-Oncology, Department of Neurology, Sandra and Edward Meyer Cancer Center, New York-Presbyterian Hospital, Weill Cornell Medicine, New York, NY, USA
| | - Stefano Cirigliano
- Sanford I. Weill Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Meyer Cancer Center, Division of Neuro-Oncology, Department of Neurology, Sandra and Edward Meyer Cancer Center, New York-Presbyterian Hospital, Weill Cornell Medicine, New York, NY, USA
| | - Syed Raza Mahmood
- Sanford I. Weill Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Uk Jin Lee
- Sanford I. Weill Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Alexander Martinez-Fundichely
- Sanford I. Weill Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA; Caryl and Israel Englander Institute for Precision Medicine, New York-Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA; Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Johannes Jungverdorben
- Center for Stem Cell Biology, Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY, USA
| | - Richa Singhania
- Sanford I. Weill Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Meyer Cancer Center, Division of Neuro-Oncology, Department of Neurology, Sandra and Edward Meyer Cancer Center, New York-Presbyterian Hospital, Weill Cornell Medicine, New York, NY, USA
| | - Sandy Rajkumar
- Sanford I. Weill Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Raphael Kirou
- Sanford I. Weill Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Lorenz Studer
- Center for Stem Cell Biology, Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY, USA
| | - Ekta Khurana
- Sanford I. Weill Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA; Caryl and Israel Englander Institute for Precision Medicine, New York-Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA; Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Alexander Polyzos
- Sanford I. Weill Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Howard A Fine
- Sanford I. Weill Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Meyer Cancer Center, Division of Neuro-Oncology, Department of Neurology, Sandra and Edward Meyer Cancer Center, New York-Presbyterian Hospital, Weill Cornell Medicine, New York, NY, USA.
| | - Effie Apostolou
- Sanford I. Weill Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA.
| |
Collapse
|
9
|
Ham DJ, Semeraro M, Berger BM, McGowan TJ, Lin S, Maino E, Oliveri F, Rüegg MA. Muscle fiber Myc is dispensable for muscle growth and its forced expression severely perturbs homeostasis. Nat Commun 2025; 16:3190. [PMID: 40180928 PMCID: PMC11968879 DOI: 10.1038/s41467-025-58542-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 03/19/2025] [Indexed: 04/05/2025] Open
Abstract
The oncogenic transcription factor Myc stimulates many growth processes including cell cycle progression and ribosome biogenesis. Myc expression is low in adult skeletal muscle, but is upregulated upon growth stimuli. Furthermore, muscle fiber Myc overexpression recapitulates many aspects of growth-related gene expression, leading to the hypothesis that Myc mediates pro-growth responses to anabolic stimuli, such as exercise. Here, we test this hypothesis by examining mouse models in which Myc is specifically eliminated or overexpressed in skeletal muscle fibers or muscle stem cells (MuSC). While muscle fiber Myc expression increased during muscle growth and Myc expression in MuSCs was required for successful muscle regeneration, muscle fiber Myc expression was dispensable for post-natal, mechanical overload or PKBα/Akt1-induced muscle growth in mice. Similarly, constitutive Myc expression did not promote skeletal muscle hypertrophy, but instead impaired muscle fiber structure and function within days. These data question the role of Myc in skeletal muscle growth.
Collapse
Affiliation(s)
- Daniel J Ham
- Biozentrum, University of Basel, Basel, Switzerland.
| | | | - Bianca M Berger
- Biozentrum, University of Basel, Basel, Switzerland
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | | | - Shuo Lin
- Biozentrum, University of Basel, Basel, Switzerland
| | | | | | | |
Collapse
|
10
|
Mahmud S, Ajadee A, Sarker A, Ahmmed R, Noor T, Pappu MAA, Islam MS, Mollah MNH. Exploring common genomic biomarkers to disclose common drugs for the treatment of colorectal cancer and hepatocellular carcinoma with type-2 diabetes through transcriptomics analysis. PLoS One 2025; 20:e0319028. [PMID: 40127075 PMCID: PMC11932495 DOI: 10.1371/journal.pone.0319028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 01/25/2025] [Indexed: 03/26/2025] Open
Abstract
Type 2 diabetes (T2D) is a crucial risk factor for both colorectal cancer (CRC) and hepatocellular carcinoma (HCC). However, so far, there was no study that has investigated common drugs against HCC and CRC during their co-occurrence with T2D patients. Consequently, patients often require multiple disease-specific multiple drugs, which can lead toxicities and adverse effects to the patients due to drug-drug interactions. This study aimed to identify common genomic biomarkers (cGBs) and associated pathogenetic mechanisms underlying CRC, HCC, and T2D to uncover potential common therapeutic compounds against these three diseases. Firstly, we identified 86 common differentially expressed genes (cDEGs) capable of separating each of CRC, HCC and T2D patients from control groups based on transcriptomic profiling. Of these cDEGs, 37 genes were upregulated and 49 were downregulated. Genetic association studies based on average of Log2 fold-change (aLog2FC) of cDEGs suggested a genetic association among CRC, HCC and T2D. Subsequently, six top-ranked cDEGs (MYC, MMP9, THBS1, IL6, CXCL1, and SPP1) were identified as common genomic biomarkers (cGBs) through protein-protein interaction (PPI) network analysis. Further analysis of these cGBs with GO-terms and KEGG pathways revealed shared pathogenetic mechanisms of three diseases, including specific biological processes, molecular functions, cellular components and signaling pathways. The gene co-regulatory network analysis identified two transcription factors (FOXC1 and GATA2) and three miRNAs (hsa-mir-195-5p, hsa-mir-124a-3p, and hsa-mir-34a-5p) as crucial transcriptional and post-transcriptional regulators of the cGBs. Finally, cGBs-guided seven candidate drugs (Digitoxin, Camptosar, AMG-900, Imatinib, Irinotecan, Midostaurin, and Linsitinib) as the common treatment against T2D, CRC and HCC were identified through molecular docking, cross-validation, and ADME/T (Absorption-Distribution-Metabolism-Excretion-Toxicity) analysis. Most of these findings received support by the literature review of diseases specific individual studies. Thus, this study offers valuable insights for researchers and clinicians to improve the diagnosis and treatment of CRC and/or HCC patients during the co-occurrence of T2D.
Collapse
Affiliation(s)
- Sabkat Mahmud
- Bioinformatics Lab (Dry), Department of Statistics, University of Rajshahi, Bangladesh
| | - Alvira Ajadee
- Bioinformatics Lab (Dry), Department of Statistics, University of Rajshahi, Bangladesh
| | - Arnob Sarker
- Bioinformatics Lab (Dry), Department of Statistics, University of Rajshahi, Bangladesh
- Department of Biochemistry & Molecular Biology, University of Rajshahi, Bangladesh
| | - Reaz Ahmmed
- Bioinformatics Lab (Dry), Department of Statistics, University of Rajshahi, Bangladesh
- Department of Biochemistry & Molecular Biology, University of Rajshahi, Bangladesh
| | - Tasfia Noor
- Department of Computer Science and Engineering, Rajshahi University of Engineering & Technology (RUET), Bangladesh
| | - Md. Al Amin Pappu
- Bioinformatics Lab (Dry), Department of Statistics, University of Rajshahi, Bangladesh
| | - Md. Saiful Islam
- Bioinformatics Lab (Dry), Department of Statistics, University of Rajshahi, Bangladesh
| | | |
Collapse
|
11
|
Cohn GM, Daniel CJ, Eng JR, Sun XX, Pelz C, Chin K, Smith A, Lopez CD, Brody JR, Dai MS, Sears RC. MYC Serine 62 phosphorylation promotes its binding to DNA double strand breaks to facilitate repair and cell survival under genotoxic stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.19.644227. [PMID: 40166231 PMCID: PMC11957152 DOI: 10.1101/2025.03.19.644227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Genomic instability is a hallmark of cancer, driving oncogenic mutations that enhance tumor aggressiveness and drug resistance. MYC, a master transcription factor that is deregulated in nearly all human tumors, paradoxically induces replication stress and associated DNA damage while also increasing expression of DNA repair factors and mediating resistance to DNA-damaging therapies. Emerging evidence supports a non-transcriptional role for MYC in preserving genomic integrity at sites of active transcription and protecting stalled replication forks under stress. Understanding how MYC's genotoxic and genoprotective functions diverge may reveal new therapeutic strategies for MYC-driven cancers. Here, we identify a non-canonical role of MYC in DNA damage response (DDR) through its direct association with DNA breaks. We show that phosphorylation at serine 62 (pS62-MYC) is crucial for the efficient recruitment of MYC to damage sites, its interaction with repair factors BRCA1 and RAD51, and effective DNA repair to support cell survival under stress. Mass spectrometry analysis with MYC-BioID2 during replication stress reveals a shift in MYC's interactome, maintaining DDR associations while losing transcriptional regulators. These findings establish pS62-MYC as a key regulator of genomic stability and a potential therapeutic target in cancers.
Collapse
Affiliation(s)
- Gabriel M. Cohn
- Department of Molecular and Medical Genetics, School of Medicine, Oregon Health and Science University, Portland, OR, USA
| | - Colin J. Daniel
- Department of Molecular and Medical Genetics, School of Medicine, Oregon Health and Science University, Portland, OR, USA
| | - Jennifer R. Eng
- Department of Molecular and Medical Genetics, School of Medicine, Oregon Health and Science University, Portland, OR, USA
| | - Xiao-Xin Sun
- Department of Molecular and Medical Genetics, School of Medicine, Oregon Health and Science University, Portland, OR, USA
| | - Carl Pelz
- Department of Molecular and Medical Genetics, School of Medicine, Oregon Health and Science University, Portland, OR, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR, USA
| | - Koei Chin
- Center for Early Detection Advanced Research, Oregon Health and Science University, Portland, OR, USA
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, USA
| | - Alexander Smith
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR, USA
| | - Charles D. Lopez
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR, USA
- Department of Hematology and Oncology, Oregon Health and Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Jonathan R. Brody
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
- Department of Surgery, Oregon Health and Science University, Portland, OR, USA
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR, USA
| | - Mu-shui Dai
- Department of Molecular and Medical Genetics, School of Medicine, Oregon Health and Science University, Portland, OR, USA
| | - Rosalie C. Sears
- Department of Molecular and Medical Genetics, School of Medicine, Oregon Health and Science University, Portland, OR, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
12
|
Komleva Y, Shpiliukova K, Bondar N, Salmina A, Khilazheva E, Illarioshkin S, Piradov M. Decoding brain aging trajectory: predictive discrepancies, genetic susceptibilities, and emerging therapeutic strategies. Front Aging Neurosci 2025; 17:1562453. [PMID: 40177249 PMCID: PMC11962000 DOI: 10.3389/fnagi.2025.1562453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 02/28/2025] [Indexed: 04/05/2025] Open
Abstract
The global extension of human lifespan has intensified the focus on aging, yet its underlying mechanisms remain inadequately understood. The article highlights aspects of genetic susceptibility to impaired brain bioenergetics, trends in age-related gene expression related to neuroinflammation and brain senescence, and the impact of stem cell exhaustion and quiescence on accelerated brain aging. We also review the accumulation of senescent cells, mitochondrial dysfunction, and metabolic disturbances as central pathological processes in aging, emphasizing how these factors contribute to inflammation and disrupt cellular competition defining the aging trajectory. Furthermore, we discuss emerging therapeutic strategies and the future potential of integrating advanced technologies to refine aging assessments. The combination of several methods including genetic analysis, neuroimaging techniques, cognitive tests and digital twins, offer a novel approach by simulating and monitoring individual health and aging trajectories, thereby providing more accurate and personalized insights. Conclusively, the accurate estimation of brain aging trajectories is crucial for understanding and managing aging processes, potentially transforming preventive and therapeutic strategies to improve health outcomes in aging populations.
Collapse
Affiliation(s)
| | | | - Nikolai Bondar
- Research Center of Neurology, Moscow, Russia
- Laboratory of Molecular Virology, First Moscow State Medical University (Sechenov University), Moscow, Russia
| | | | - Elena Khilazheva
- Department of Biological Chemistry with Courses in Medical, Research Institute of Molecular Medicine and Pathobiochemistry, Pharmaceutical and Toxicological Chemistry Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University of the Ministry of Healthcare of the Russian Federation, Krasnoyarsk, Russia
| | | | | |
Collapse
|
13
|
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] [Download PDF] [Figures] [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
|
14
|
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] [Download PDF] [Figures] [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
|
15
|
Lin F, Zheng WC, Ke ZB, Chen DN, Xue YT, Lin YZ, Li XD, Zheng QS, Wei Y, Xue XY, Xu N. A comprehensive analysis-based study of Di-(2-ethylhexyl) phthalate (DEHP)-Environmental explanation of bladder cancer progression. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 367:125625. [PMID: 39746639 DOI: 10.1016/j.envpol.2024.125625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 12/28/2024] [Accepted: 12/31/2024] [Indexed: 01/04/2025]
Abstract
Di-(2-ethylhexyl) phthalate (DEHP) is widely utilized as a plasticizer in industrial manufacturing to enhance the durability and flexibility of plastics. Studies have depicted that DEHP exposure may be associated with multiple cancers, including colorectal, liver and prostate cancer. However, the effects and underlying mechanisms of DEHP on bladder cancer progression remain unspecific. In this work, we explored the correlation between DEHP exposure and bladder cancer through comprehensive analysis. A total of 172 differentially expressed DEHP-related genes (DEDRGs) were screened based on the CTD and TCGA database. In addition, a new prognostic model related to DEHP was developed, which had excellent predictive power for bladder cancer prognoses. Molecular docking techniques were employed to assess the binding affinity of mono(2-ethylhexyl) phthalate (MEHP) to crucial proteins. Lastly, both in vitro and in vivo experiments, along with RNA sequencing, were conducted to elucidate the biological roles and mechanisms of MEHP in bladder cancer. Several new insights into the role of DEHP/MEHP in bladder cancer were given in this study, as well as an awareness of the association between environmental toxicants and cancer progression.
Collapse
Affiliation(s)
- Fei Lin
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China; Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Wen-Cai Zheng
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China; Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Zhi-Bin Ke
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China; Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Dong-Ning Chen
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China; Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Yu-Ting Xue
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China; Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Yun-Zhi Lin
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China; Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Xiao-Dong Li
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China; Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Qing-Shui Zheng
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China; Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Yong Wei
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China; Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Xue-Yi Xue
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China; Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China; Fujian Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
| | - Ning Xu
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China; Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China; Fujian Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
| |
Collapse
|
16
|
Edaibis R, Akel R, Shin JA. Beyond small molecules: advancing MYC-targeted cancer therapies through protein engineering. Transcription 2025; 16:67-85. [PMID: 39878458 PMCID: PMC11970745 DOI: 10.1080/21541264.2025.2453315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 01/31/2025] Open
Abstract
Protein engineering has emerged as a powerful approach toward the development of novel therapeutics targeting the MYC/MAX/E-box network, an active driver of >70% of cancers. The MYC/MAX heterodimer regulates numerous genes in our cells by binding the Enhancer box (E-box) DNA site and activating the transcription of downstream genes. Traditional small molecules that inhibit MYC face significant limitations that include toxic effects, drug delivery challenges, and resistance. Recent advances in protein engineering offer promising alternatives by creating protein-based drugs that directly disrupt the MYC/MAX dimerization interface and/or MYC/MAX's binding to specific DNA targets. Designed DNA binding proteins like Omomyc, DuoMyc, ME47, MEF, and Mad inhibit MYC activity through specific dimerization, sequestration, and DNA-binding mechanisms. Compared to small molecules, these engineered proteins can offer superior specificity and efficacy and provide a potential pathway for overcoming the limitations of traditional cancer therapies. The success of these protein therapeutics highlights the importance of protein engineering in developing cancer treatments.
Collapse
Affiliation(s)
- Rama Edaibis
- Department of Chemistry, University of Toronto, Mississauga, ON, Canada
| | - Raneem Akel
- Department of Chemistry, University of Toronto, Mississauga, ON, Canada
| | - Jumi A. Shin
- Department of Chemistry, University of Toronto, Mississauga, ON, Canada
| |
Collapse
|
17
|
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
|
18
|
Breves SL, Di Giammartino DC, Nicholson J, Cirigliano S, Mahmood SR, Lee UJ, Martinez-Fundichely A, Jungverdorben J, Singhania R, Rajkumar S, Kirou R, Studer L, Khurana E, Polyzos A, Fine HA, Apostolou E. Three-dimensional regulatory hubs support oncogenic programs in glioblastoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.20.629544. [PMID: 40034649 PMCID: PMC11875237 DOI: 10.1101/2024.12.20.629544] [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: 03/05/2025]
Abstract
Dysregulation of enhancer-promoter communication in the context of the three-dimensional (3D) nucleus is increasingly recognized as a potential driver of oncogenic programs. Here, we profiled the 3D enhancer-promoter networks of primary patient-derived glioblastoma stem cells (GSCs) in comparison with neuronal stem cells (NSCs) to identify potential central nodes and vulnerabilities in the regulatory logic of this devastating cancer. Specifically, we focused on hyperconnected 3D regulatory hubs and demonstrated that hub-interacting genes exhibit high and coordinated expression at the single-cell level and strong association with oncogenic programs that distinguish IDH-wt glioblastoma patients from low-grade glioma. Epigenetic silencing of a recurrent 3D enhancer hub-with an uncharacterized role in glioblastoma-was sufficient to cause concordant downregulation of multiple hub-connected genes along with significant shifts in transcriptional states and reduced clonogenicity. By integrating published datasets from other cancer types, we also identified both universal and cancer type-specific 3D regulatory hubs which enrich for varying oncogenic programs and nominate specific factors associated with worse outcomes. Genetic alterations, such as focal duplications, could explain only a small fraction of the detected hyperconnected hubs and their increased activity. Overall, our study provides computational and experimental support for the potential central role of 3D regulatory hubs in controlling oncogenic programs and properties.
Collapse
Affiliation(s)
- Sarah L. Breves
- Sanford I. Weill Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, United States
- Physiology, Biophysics and Systems Biology Program, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY, USA
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Dafne Campigli Di Giammartino
- Sanford I. Weill Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, United States
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
- 3D Chromatin Conformation and RNA genomics laboratory, Istituto Italiano di Tecnologia (IIT), Center for Human Technologies (CHT), Genova, Italy (current affiliation)
| | - James Nicholson
- Meyer Cancer Center, Division of Neuro-Oncology, Department of Neurology, Sandra and Edward Meyer Cancer Center, New York-Presbyterian Hospital/Weill Cornell Medicine, New York, NY, USA
| | - Stefano Cirigliano
- Meyer Cancer Center, Division of Neuro-Oncology, Department of Neurology, Sandra and Edward Meyer Cancer Center, New York-Presbyterian Hospital/Weill Cornell Medicine, New York, NY, USA
| | - Syed Raza Mahmood
- Sanford I. Weill Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, United States
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Uk Jin Lee
- Sanford I. Weill Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, United States
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Alexander Martinez-Fundichely
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, 10021, USA
- Meyer Cancer Center, Sandra and Edward Meyer Cancer Center, New York-Presbyterian Hospital/Weill Cornell Medicine, New York, NY, USA
- Caryl and Israel Englander Institute for Precision Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Johannes Jungverdorben
- The Center for Stem Cell Biology, Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY, USA
| | - Richa Singhania
- Meyer Cancer Center, Division of Neuro-Oncology, Department of Neurology, Sandra and Edward Meyer Cancer Center, New York-Presbyterian Hospital/Weill Cornell Medicine, New York, NY, USA
| | - Sandy Rajkumar
- Sanford I. Weill Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, United States
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Raphael Kirou
- Sanford I. Weill Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, United States
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Lorenz Studer
- The Center for Stem Cell Biology, Developmental Biology Program, Sloan Kettering Institute for Cancer Research, New York, NY, USA
| | - Ekta Khurana
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, 10021, USA
- Meyer Cancer Center, Sandra and Edward Meyer Cancer Center, New York-Presbyterian Hospital/Weill Cornell Medicine, New York, NY, USA
- Caryl and Israel Englander Institute for Precision Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Alexander Polyzos
- Sanford I. Weill Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, United States
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Howard A. Fine
- 3D Chromatin Conformation and RNA genomics laboratory, Istituto Italiano di Tecnologia (IIT), Center for Human Technologies (CHT), Genova, Italy (current affiliation)
| | - Effie Apostolou
- Sanford I. Weill Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, United States
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| |
Collapse
|
19
|
Guiyedi K, Parquet M, Aoufouchi S, Chauzeix J, Rizzo D, Al Jamal I, Feuillard J, Gachard N, Peron S. Increased c-MYC Expression Associated with Active IGH Locus Rearrangement: An Emerging Role for c-MYC in Chronic Lymphocytic Leukemia. Cancers (Basel) 2024; 16:3749. [PMID: 39594704 PMCID: PMC11592262 DOI: 10.3390/cancers16223749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 10/25/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
This review examines the pivotal role of c-MYC in Chronic Lymphocytic Leukemia (CLL), focusing on how its overexpression leads to increased genetic instability, thereby accelerating disease progression. MYC, a major oncogene, encodes a transcription factor that regulates essential cellular processes, including cell cycle control, proliferation, and apoptosis. In CLL cases enriched with unmutated immunoglobulin heavy chain variable (IGHV) genes, MYC is significantly overexpressed and associated with active rearrangements in the IGH immunoglobulin heavy chain locus. This overexpression results in substantial DNA damage, including double-strand breaks, chromosomal translocations, and an increase in abnormal repair events. Consequently, c-MYC plays a dual role in CLL: it promotes aggressive cell proliferation while concurrently driving genomic instability through its involvement in genetic recombination. This dynamic contributes not only to CLL progression but also to the overall aggressiveness of the disease. Additionally, the review suggests that c-MYC's influence on genetic rearrangements makes it an attractive target for therapeutic strategies aimed at mitigating CLL malignancy. These findings underscore c-MYC's critical importance in advancing CLL progression, highlighting the need for further research to explore its potential as a target in future treatment approaches.
Collapse
Affiliation(s)
- Kenza Guiyedi
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 7276/INSERM U1262, Université de Limoges, 87000 Limoges, France
| | - Milène Parquet
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 7276/INSERM U1262, Université de Limoges, 87000 Limoges, France
| | - Said Aoufouchi
- Gustave Roussy, B-Cell and Genome Plasticity Team, CNRS UMR9019, Villejuif, France and Université Paris-Saclay, 91400 Orsay, France
| | - Jasmine Chauzeix
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 7276/INSERM U1262, Université de Limoges, 87000 Limoges, France
- Laboratoire d’Hématologie Biologique, Centre Hospitalier Universitaire de Limoges, 87000 Limoges, France
| | - David Rizzo
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 7276/INSERM U1262, Université de Limoges, 87000 Limoges, France
- Laboratoire d’Hématologie Biologique, Centre Hospitalier Universitaire de Limoges, 87000 Limoges, France
| | - Israa Al Jamal
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 7276/INSERM U1262, Université de Limoges, 87000 Limoges, France
- Faculty of Sciences, GSBT Genomic Surveillance and Biotherapy Team, Mont Michel Campus, Lebanese University, Tripoli 1300, Lebanon
| | - Jean Feuillard
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 7276/INSERM U1262, Université de Limoges, 87000 Limoges, France
- Laboratoire d’Hématologie Biologique, Centre Hospitalier Universitaire de Limoges, 87000 Limoges, France
| | - Nathalie Gachard
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 7276/INSERM U1262, Université de Limoges, 87000 Limoges, France
- Laboratoire d’Hématologie Biologique, Centre Hospitalier Universitaire de Limoges, 87000 Limoges, France
| | - Sophie Peron
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR) 7276/INSERM U1262, Université de Limoges, 87000 Limoges, France
| |
Collapse
|
20
|
Giacomini A, Taranto S, Gazzaroli G, Faletti J, Capoferri D, Marcheselli R, Sciumè M, Presta M, Sacco A, Roccaro AM. The FGF/FGFR/c-Myc axis as a promising therapeutic target in multiple myeloma. J Exp Clin Cancer Res 2024; 43:294. [PMID: 39482742 PMCID: PMC11529022 DOI: 10.1186/s13046-024-03217-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/26/2024] [Indexed: 11/03/2024] Open
Abstract
Among blood cancers, multiple myeloma (MM) represents the second most common neoplasm and is characterized by the accumulation and proliferation of monoclonal plasma cells within the bone marrow. Despite the last few decades being characterized by the development of different therapeutic strategies against MM, at present such disease is still considered incurable. Although MM is highly heterogeneous in terms of genetic and molecular subtypes, about 67% of MM cases are associated with abnormal activity of the transcription factor c-Myc, which has so far revealed a protein extremely difficult to target. We have recently demonstrated that activation of fibroblast growth factor (FGF) signaling protects MM cells from oxidative stress-induced apoptosis by stabilizing the oncoprotein c-Myc. Accordingly, secretion of FGF ligands and autocrine activation of FGF receptors (FGFR) is observed in MM cells and FGFR3 genomic alterations represent some 15-20% MM cases and are associated with poor outcome. Thus, FGF/FGFR blockade may represent a promising strategy to indirectly target c-Myc in MM. On this basis, the present review aims at providing an overview of recently explored connections between the FGF/FGFR system and c-Myc oncoprotein, sustaining the therapeutic potential of targeting the FGF/FGFR/c-Myc axis in MM by using inhibitors targeting FGF ligands or FGF receptors. Importantly, the provided findings may represent the rationale for using FDA approved FGFR TK inhibitors (i.e. Pemigatinib, Futibatinib, Erdafitinib) for the treatment of MM patients presenting with an aberrant activation of this axis.
Collapse
Affiliation(s)
- Arianna Giacomini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| | - Sara Taranto
- Clinical Trial Center, Translational Research and Phase I Unit, ASST Spedali Civili Di Brescia, Brescia, Italy
| | - Giorgia Gazzaroli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Jessica Faletti
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Davide Capoferri
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Raffaella Marcheselli
- Clinical Trial Center, Translational Research and Phase I Unit, ASST Spedali Civili Di Brescia, Brescia, Italy
| | - Margherita Sciumè
- Clinical Trial Center, Translational Research and Phase I Unit, ASST Spedali Civili Di Brescia, Brescia, Italy
| | - Marco Presta
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Antonio Sacco
- Clinical Trial Center, Translational Research and Phase I Unit, ASST Spedali Civili Di Brescia, Brescia, Italy
| | - Aldo M Roccaro
- Clinical Trial Center, Translational Research and Phase I Unit, ASST Spedali Civili Di Brescia, Brescia, Italy.
| |
Collapse
|
21
|
Sevrin T, Imoto H, Robertson S, Rauch N, Dyn'ko U, Koubova K, Wynne K, Kolch W, Rukhlenko OS, Kholodenko BN. Cell-specific models reveal conformation-specific RAF inhibitor combinations that synergistically inhibit ERK signaling in pancreatic cancer cells. Cell Rep 2024; 43:114710. [PMID: 39240715 PMCID: PMC11474227 DOI: 10.1016/j.celrep.2024.114710] [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: 12/19/2023] [Revised: 07/16/2024] [Accepted: 08/20/2024] [Indexed: 09/08/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) presents significant challenges for targeted clinical interventions due to prevalent KRAS mutations, rendering PDAC resistant to RAF and MEK inhibitors (RAFi and MEKi). In addition, responses to targeted therapies vary between patients. Here, we explored the differential sensitivities of PDAC cell lines to RAFi and MEKi and developed an isogenic pair comprising the most sensitive and resistant PDAC cells. To simulate patient- or tumor-specific variations, we constructed cell-line-specific mechanistic models based on protein expression profiling and differential properties of KRAS mutants. These models predicted synergy between two RAFi with different conformation specificity (type I½ and type II RAFi) in inhibiting phospho-ERK (ppERK) and reducing PDAC cell viability. This synergy was experimentally validated across all four studied PDAC cell lines. Our findings underscore the need for combination approaches to inhibit the ERK pathway in PDAC.
Collapse
Affiliation(s)
- Thomas Sevrin
- Systems Biology Ireland, University College Dublin, Dublin, Ireland
| | - Hiroaki Imoto
- Systems Biology Ireland, University College Dublin, Dublin, Ireland
| | - Sarah Robertson
- Systems Biology Ireland, University College Dublin, Dublin, Ireland
| | - Nora Rauch
- Systems Biology Ireland, University College Dublin, Dublin, Ireland
| | - Uscinnia Dyn'ko
- Systems Biology Ireland, University College Dublin, Dublin, Ireland
| | - Katerina Koubova
- Systems Biology Ireland, University College Dublin, Dublin, Ireland; Department of Histology and Embryology, Faculty of Medicine and Dentistry, Palacky University, 779 00 Olomouc, Czech Republic
| | - Kieran Wynne
- Systems Biology Ireland, University College Dublin, Dublin, Ireland
| | - Walter Kolch
- Systems Biology Ireland, University College Dublin, Dublin, Ireland; Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland; School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| | | | - Boris N Kholodenko
- Systems Biology Ireland, University College Dublin, Dublin, Ireland; Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland; School of Medicine and Medical Science, University College Dublin, Dublin, Ireland; Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
22
|
Di Pietro B, Villata S, Dal Monego S, Degasperi M, Ghini V, Guarnieri T, Plaksienko A, Liu Y, Pecchioli V, Manni L, Tenori L, Licastro D, Angelini C, Napione L, Frascella F, Nardini C. Differential Anti-Inflammatory Effects of Electrostimulation in a Standardized Setting. Int J Mol Sci 2024; 25:9808. [PMID: 39337300 PMCID: PMC11432240 DOI: 10.3390/ijms25189808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/28/2024] [Accepted: 09/03/2024] [Indexed: 09/30/2024] Open
Abstract
The therapeutic usage of physical stimuli is framed in a highly heterogeneous research area, with variable levels of maturity and of translatability into clinical application. In particular, electrostimulation is deeply studied for its application on the autonomous nervous system, but less is known about the anti- inflammatory effects of such stimuli beyond the inflammatory reflex. Further, reproducibility and meta-analyses are extremely challenging, owing to the limited rationale on dosage and experimental standardization. It is specifically to address the fundamental question on the anti-inflammatory effects of electricity on biological systems, that we propose a series of controlled experiments on the effects of direct and alternate current delivered on a standardized 3D bioconstruct constituted by fibroblasts and keratinocytes in a collagen matrix, in the presence or absence of TNF-α as conventional inflammation inducer. This selected but systematic exploration, with transcriptomics backed by metabolomics at specific time points allows to obtain the first systemic overview of the biological functions at stake, highlighting the differential anti-inflammatory potential of such approaches, with promising results for 5 V direct current stimuli, correlating with the wound healing process. With our results, we wish to set the base for a rigorous systematic approach to the problem, fundamental towards future elucidations of the detailed mechanisms at stake, highlighting both the healing and damaging potential of such approaches.
Collapse
Affiliation(s)
- Biagio Di Pietro
- Consiglio Nazionale delle Ricerche, Istituto per le Applicazioni del Calcolo “Mauro Picone”, 00185 Roma, Italy; (B.D.P.); (T.G.); (A.P.); (Y.L.); (C.A.)
| | - Simona Villata
- Dipartimento di Scienza Applicata e Tecnologia, Politecnico di Torino, 10129 Turin, Italy; (S.V.); (F.F.)
- PolitoBIOMed Lab, Politecnico di Torino, 10129 Turin, Italy
| | - Simeone Dal Monego
- Area Science Park, Basovizza, 34149 Trieste, Italy; (S.D.M.); (M.D.); (D.L.)
| | | | - Veronica Ghini
- Department of Chemistry “Ugo Schiff” and Magnetic Resonance Center (CERM), University of Florence, 50019 Sesto Fiorentino, Italy; (V.G.); (L.T.)
| | - Tiziana Guarnieri
- Consiglio Nazionale delle Ricerche, Istituto per le Applicazioni del Calcolo “Mauro Picone”, 00185 Roma, Italy; (B.D.P.); (T.G.); (A.P.); (Y.L.); (C.A.)
- Dipartimento di Scienze Biologiche, Geologiche e Ambientali (BIGEA), University of Bologna, 40100 Bologna, Italy
| | - Anna Plaksienko
- Consiglio Nazionale delle Ricerche, Istituto per le Applicazioni del Calcolo “Mauro Picone”, 00185 Roma, Italy; (B.D.P.); (T.G.); (A.P.); (Y.L.); (C.A.)
- Oslo Center of Biostatistics and Epidemiology, University of Oslo, 0317 Oslo, Norway
| | - Yuanhua Liu
- Consiglio Nazionale delle Ricerche, Istituto per le Applicazioni del Calcolo “Mauro Picone”, 00185 Roma, Italy; (B.D.P.); (T.G.); (A.P.); (Y.L.); (C.A.)
- State Key Laboratory of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Valentina Pecchioli
- Consorzio Interuniversitario Risonanze Magnetiche Metallo Proteine (CIRMMP), 50019 Sesto Fiorentino, Italy;
| | - Luigi Manni
- Consiglio Nazionale delle Ricerche, Istituto di Farmacologia Traslazionale, 00185 Roma, Italy;
| | - Leonardo Tenori
- Department of Chemistry “Ugo Schiff” and Magnetic Resonance Center (CERM), University of Florence, 50019 Sesto Fiorentino, Italy; (V.G.); (L.T.)
- Consorzio Interuniversitario Risonanze Magnetiche Metallo Proteine (CIRMMP), 50019 Sesto Fiorentino, Italy;
| | - Danilo Licastro
- Area Science Park, Basovizza, 34149 Trieste, Italy; (S.D.M.); (M.D.); (D.L.)
| | - Claudia Angelini
- Consiglio Nazionale delle Ricerche, Istituto per le Applicazioni del Calcolo “Mauro Picone”, 00185 Roma, Italy; (B.D.P.); (T.G.); (A.P.); (Y.L.); (C.A.)
| | - Lucia Napione
- Dipartimento di Scienza Applicata e Tecnologia, Politecnico di Torino, 10129 Turin, Italy; (S.V.); (F.F.)
- PolitoBIOMed Lab, Politecnico di Torino, 10129 Turin, Italy
| | - Francesca Frascella
- Dipartimento di Scienza Applicata e Tecnologia, Politecnico di Torino, 10129 Turin, Italy; (S.V.); (F.F.)
- PolitoBIOMed Lab, Politecnico di Torino, 10129 Turin, Italy
| | - Christine Nardini
- Consiglio Nazionale delle Ricerche, Istituto per le Applicazioni del Calcolo “Mauro Picone”, 00185 Roma, Italy; (B.D.P.); (T.G.); (A.P.); (Y.L.); (C.A.)
| |
Collapse
|
23
|
Ormsbee Golden BD, Gonzalez DV, Yochum GS, Coulter DW, Rizzino A. SOX2 represses c-MYC transcription by altering the co-activator landscape of the c-MYC super-enhancer and promoter regions. J Biol Chem 2024; 300:107642. [PMID: 39122009 PMCID: PMC11408076 DOI: 10.1016/j.jbc.2024.107642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 07/05/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Our previous studies determined that elevating SOX2 in a wide range of tumor cells leads to a reversible state of tumor growth arrest. Efforts to understand how tumor cell growth is inhibited led to the discovery of a SOX2:MYC axis that is responsible for downregulating c-MYC (MYC) when SOX2 is elevated. Although we had determined that elevating SOX2 downregulates MYC transcription, the mechanism responsible was not determined. Given the challenges of targeting MYC clinically, we set out to identify how elevating SOX2 downregulates MYC transcription. In this study, we focused on the MYC promoter region and an upstream region of the MYC locus that contains a MYC super-enhancer encompassing five MYC enhancers and which is associated with several cancers. Here we report that BRD4 and p300 associate with each of the MYC enhancers in the upstream MYC super-enhancer as well as the MYC promoter region and that elevating SOX2 decreases the recruitment of BRD4 and p300 to these sites. Additionally, we determined that elevating SOX2 leads to increases in the association of SOX2 and H3K27me3 within the MYC super-enhancer and the promoter region of MYC. Importantly, we conclude that the increases in SOX2 within the MYC super-enhancer precipitate a cascade of events that culminates in the repression of MYC transcription. Together, our studies identify a novel molecular mechanism able to regulate MYC transcription in two distinctly different tumor types and provide new mechanistic insights into the molecular interrelationships between two master regulators, SOX2 and MYC, widely involved in multiple cancers.
Collapse
Affiliation(s)
- Briana D Ormsbee Golden
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Daisy V Gonzalez
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Gregory S Yochum
- Department of Surgery & Biochemistry & Molecular Biology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Donald W Coulter
- Hematology and Oncology Division, Department of Pediatrics, Nebraska Medical Center, Omaha, Nebraska, USA; Child Health Research Institute, University of Nebraska Medical Center, Omaha, Nebraska, USA; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Angie Rizzino
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, USA.
| |
Collapse
|
24
|
Luković D, Franich AA, Živković MD, Rajković S, Stojanović B, Gajović N, Jurišević M, Pavlović S, Simović Marković B, Jovanović M, Stojanović BS, Pavlović R, Jovanović I. Biological Evaluation of Dinuclear Platinum(II) Complexes with Aromatic N-Heterocycles as Bridging Ligands. Int J Mol Sci 2024; 25:8525. [PMID: 39126093 PMCID: PMC11312983 DOI: 10.3390/ijms25158525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024] Open
Abstract
The history of effective anti-cancer medications begins with the discovery of cisplatin's anti-cancer properties. Second-generation analogue, carboplatin, with a similar range of effectiveness, made progress in improving these drugs with fewer side effects and better solubility. Renewed interest in platinum-based drugs has been increasing in the past several years. These developments highlight a revitalized enthusiasm and ongoing exploration in platinum chemotherapy based on the series of dinuclear platinum(II) complexes, [{Pt(L)Cl}2(μ-bridging ligand)]2+, which have been synthesized and evaluated for their biological activities. These complexes are designed to target various cancerous conditions, exhibiting promising antitumor, antiproliferative, and apoptosis-inducing activities. The current work aims to shed light on the potential of these complexes as next-generation platinum-based therapies, highlighting their enhanced efficacy and reduced side effects, which could revolutionize the approach to chemotherapy.
Collapse
Affiliation(s)
- Desimir Luković
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, S. Markovića 69, 34000 Kragujevac, Serbia; (D.L.); (N.G.); (M.J.); (S.P.); (B.S.M.); (M.J.); (B.S.S.); (I.J.)
| | - Andjela A. Franich
- Department of Chemistry, Faculty of Science, University of Kragujevac, R. Domanovića 12, 34000 Kragujevac, Serbia; (A.A.F.); (S.R.)
| | - Marija D. Živković
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, S. Markovića 69, 34000 Kragujevac, Serbia
| | - Snežana Rajković
- Department of Chemistry, Faculty of Science, University of Kragujevac, R. Domanovića 12, 34000 Kragujevac, Serbia; (A.A.F.); (S.R.)
| | - Bojan Stojanović
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, S. Markovića 69, 34000 Kragujevac, Serbia; (D.L.); (N.G.); (M.J.); (S.P.); (B.S.M.); (M.J.); (B.S.S.); (I.J.)
- Department of Surgery, Faculty of Medical Sciences, University of Kragujevac, S. Markovića 69, 34000 Kragujevac, Serbia
| | - Nevena Gajović
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, S. Markovića 69, 34000 Kragujevac, Serbia; (D.L.); (N.G.); (M.J.); (S.P.); (B.S.M.); (M.J.); (B.S.S.); (I.J.)
| | - Milena Jurišević
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, S. Markovića 69, 34000 Kragujevac, Serbia; (D.L.); (N.G.); (M.J.); (S.P.); (B.S.M.); (M.J.); (B.S.S.); (I.J.)
- Department of Clinical Pharmacy, Faculty of Medical Sciences, University of Kragujevac, S. Markovića 69, 34000 Kragujevac, Serbia;
| | - Slađana Pavlović
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, S. Markovića 69, 34000 Kragujevac, Serbia; (D.L.); (N.G.); (M.J.); (S.P.); (B.S.M.); (M.J.); (B.S.S.); (I.J.)
| | - Bojana Simović Marković
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, S. Markovića 69, 34000 Kragujevac, Serbia; (D.L.); (N.G.); (M.J.); (S.P.); (B.S.M.); (M.J.); (B.S.S.); (I.J.)
| | - Marina Jovanović
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, S. Markovića 69, 34000 Kragujevac, Serbia; (D.L.); (N.G.); (M.J.); (S.P.); (B.S.M.); (M.J.); (B.S.S.); (I.J.)
| | - Bojana S. Stojanović
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, S. Markovića 69, 34000 Kragujevac, Serbia; (D.L.); (N.G.); (M.J.); (S.P.); (B.S.M.); (M.J.); (B.S.S.); (I.J.)
- Department of Pathophysiology, Faculty of Medical Sciences, University of Kragujevac, S. Markovića 69, 34000 Kragujevac, Serbia
| | - Radiša Pavlović
- Department of Clinical Pharmacy, Faculty of Medical Sciences, University of Kragujevac, S. Markovića 69, 34000 Kragujevac, Serbia;
| | - Ivan Jovanović
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, S. Markovića 69, 34000 Kragujevac, Serbia; (D.L.); (N.G.); (M.J.); (S.P.); (B.S.M.); (M.J.); (B.S.S.); (I.J.)
| |
Collapse
|
25
|
Jiang W, Wu Y, Pang A, Li P, Mei S. M1-type microglia-derived exosomes contribute to blood-brain barrier damage. Brain Res 2024; 1835:148919. [PMID: 38588846 DOI: 10.1016/j.brainres.2024.148919] [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: 02/26/2024] [Revised: 04/02/2024] [Accepted: 04/05/2024] [Indexed: 04/10/2024]
Abstract
BACKGROUND As a key substance for intercellular communication, exosomes could be a potential strategy for stroke treatment. Activated microglia disrupt the integrity of blood-brain barrier (BBB) to facilitate the stroke process. Hence, this study was designed to investigate the effect of microglia-derived exosomes on BBB cell model injury and to explore the underlying molecular mechanisms. METHODS M1 polarization of BV2 cells was induced with LPS and their derived exosomes were isolated. Astrocytes were cultured in primary culture and constructed with End3 cells as a BBB cell model. After co-culture with exosomes, the BBB cell model was examined for changes in TEER, permeability, and expression of BBB-related proteins (Claudin-1, Occludin, ZO-1 and JAM). Resting and M1-type BV2 cell-derived exosomes perform small RNA sequences and differentially expressed miRNAs (DE-miRNAs) are identified by bioinformatics. RESULTS M1-type BV2 cell-derived exosomes decreased End3 cell viability, and increased their apoptotic ratio. Moreover, M1 type BV2 cell-derived exosomes dramatically enhanced the permeability of BBB cell model, and diminished the TEER and BBB-related protein (Claudin-1, Occludin, ZO-1) expression. Notably, resting BV2 cell-derived exosomes had no effect on the integrity of BBB cell model. Sequencing results indicated that 71 DE-miRNAs were present in M1 BV2 cell-derived exosomes, and their targets mediated neurological development and signaling pathways such as MAPK and cAMP. RT-qPCR confirmed the differential expression of mmu-miR-125a-5p, mmu-miR-122b-3p, mmu-miR-139-3p, mmu-miR-330-3p, mmu-miR-3057-5p and mmu-miR-342-3p consistent with the small RNA sequence. Furthermore, Creb1, Jun, Mtor, Frk, Pabpc1 and Sdc1 are the most well-connected proteins in the PPI network. CONCLUSION M1-type microglia-derived exosomes contribute to the injury of BBB cell model, which has the involvement of miRNAs. Our findings provide new perspectives and potential mechanisms for future M1 microglia-derived exosomes as therapeutic targets in stroke.
Collapse
Affiliation(s)
- Wen Jiang
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, No.295 Xichang Road, Kunming 650032, Yunnan, China; The Yunnan Province Clinical Research Center for Neurological Diseases, No.295 Xichang Road, Kunming 650032, Yunnan, China
| | - Yan Wu
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, No.295 Xichang Road, Kunming 650032, Yunnan, China
| | - Ailan Pang
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, No.295 Xichang Road, Kunming 650032, Yunnan, China
| | - Peiyao Li
- Department of Pain Medicine, the First Affiliated Hospital of Kunming Medical University, No.295 Xichang Road, Kunming 650032, Yunnan, China
| | - Song Mei
- Department of Cardiac Surgery, the First Affiliated Hospital of Kunming Medical University, No.295 Xichang Road, Kunming 650032, Yunnan, China.
| |
Collapse
|
26
|
Ascanelli C, Dahir R, Wilson CH. Manipulating Myc for reparative regeneration. Front Cell Dev Biol 2024; 12:1357589. [PMID: 38577503 PMCID: PMC10991803 DOI: 10.3389/fcell.2024.1357589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 02/15/2024] [Indexed: 04/06/2024] Open
Abstract
The Myc family of proto-oncogenes is a key node for the signal transduction of external pro-proliferative signals to the cellular processes required for development, tissue homoeostasis maintenance, and regeneration across evolution. The tight regulation of Myc synthesis and activity is essential for restricting its oncogenic potential. In this review, we highlight the central role that Myc plays in regeneration across the animal kingdom (from Cnidaria to echinoderms to Chordata) and how Myc could be employed to unlock the regenerative potential of non-regenerative tissues in humans for therapeutic purposes. Mastering the fine balance of harnessing the ability of Myc to promote transcription without triggering oncogenesis may open the door to many exciting opportunities for therapeutic development across a wide array of diseases.
Collapse
Affiliation(s)
| | | | - Catherine H. Wilson
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
27
|
Kargbo RB. AI in Pharma: Transforming Drug Discovery and Strategic Management with MYC-Modulating Compounds and BET Protein Inhibitors. ACS Med Chem Lett 2024; 15:334-336. [PMID: 38505845 PMCID: PMC10945536 DOI: 10.1021/acsmedchemlett.4c00057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Indexed: 03/21/2024] Open
Abstract
The landscape of pharmaceutical R&D is being reshaped by the synergistic integration of Artificial Intelligence (AI) and groundbreaking drug discoveries, mainly focusing on MYC-modulating compounds and BET protein inhibitors. This Patent Highlight delves into this convergence, illustrating a transformative shift in the pharmaceutical industry's approach to drug development, strategic management, and treating various diseases, from cancer to inflammatory and fibrotic disorders.
Collapse
|
28
|
Zacarías-Fluck MF, Soucek L, Whitfield JR. MYC: there is more to it than cancer. Front Cell Dev Biol 2024; 12:1342872. [PMID: 38510176 PMCID: PMC10952043 DOI: 10.3389/fcell.2024.1342872] [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: 11/22/2023] [Accepted: 02/20/2024] [Indexed: 03/22/2024] Open
Abstract
MYC is a pleiotropic transcription factor involved in multiple cellular processes. While its mechanism of action and targets are not completely elucidated, it has a fundamental role in cellular proliferation, differentiation, metabolism, ribogenesis, and bone and vascular development. Over 4 decades of research and some 10,000 publications linking it to tumorigenesis (by searching PubMed for "MYC oncogene") have led to MYC becoming a most-wanted target for the treatment of cancer, where many of MYC's physiological functions become co-opted for tumour initiation and maintenance. In this context, an abundance of reviews describes strategies for potentially targeting MYC in the oncology field. However, its multiple roles in different aspects of cellular biology suggest that it may also play a role in many additional diseases, and other publications are indeed linking MYC to pathologies beyond cancer. Here, we review these physiological functions and the current literature linking MYC to non-oncological diseases. The intense efforts towards developing MYC inhibitors as a cancer therapy will potentially have huge implications for the treatment of other diseases. In addition, with a complementary approach, we discuss some diseases and conditions where MYC appears to play a protective role and hence its increased expression or activation could be therapeutic.
Collapse
Affiliation(s)
- Mariano F. Zacarías-Fluck
- Models of Cancer Therapies Laboratory, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Laura Soucek
- Models of Cancer Therapies Laboratory, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Peptomyc S.L., Barcelona, Spain
| | - Jonathan R. Whitfield
- Models of Cancer Therapies Laboratory, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| |
Collapse
|
29
|
Zhang W, Wang R, Yi Z, Guo R, Li Y, Xu Y, Li X, Song J. Investigation of the Expression and Regulation of SCG5 in the Context of the Chromogranin-Secretogranin Family in Malignant Tumors. Protein Pept Lett 2024; 31:657-666. [PMID: 39219421 DOI: 10.2174/0109298665325956240819064853] [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: 06/05/2024] [Revised: 07/15/2024] [Accepted: 07/24/2024] [Indexed: 09/04/2024]
Abstract
The SCG5 gene has been demonstrated to play an essential role in the development and progression of a range of malignant neoplasms. The regulation of SCG5 expression involves multiple biological pathways. According to relevant studies, SCG5 is differentially expressed in different cancers, and its up- or down-regulation may even affect tumour growth, invasion, and migration, which caught our attention. Therefore, we summarise the regulatory roles played by the SCG5 gene in a variety of cancers and the biological regulatory mechanisms associated with its possible promotion or inhibition of tumour biological behavior, to further explore the potential of SCG5 as a new tumour marker and hopefully provide theoretical guidance for subsequent disease research and treatment.
Collapse
Affiliation(s)
- Weisong Zhang
- Department of Thoracic Surgery, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People's Hospital, Yancheng 224000, P.R. China
- Medical School of Nantong University, Nantong, 226007, P.R. China
| | - Rui Wang
- Department of Thoracic Surgery, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People's Hospital, Yancheng 224000, P.R. China
- Medical School of Nantong University, Nantong, 226007, P.R. China
| | - Zhongquan Yi
- Central laboratory, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People's Hospital, Yancheng 224000, P.R. China
| | - Rongqi Guo
- Department of Thoracic Surgery, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People's Hospital, Yancheng 224000, P.R. China
- Medical School of Nantong University, Nantong, 226007, P.R. China
| | - Yangyang Li
- Department of Thoracic Surgery, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People's Hospital, Yancheng 224000, P.R. China
- Medical School of Nantong University, Nantong, 226007, P.R. China
| | - Yanhan Xu
- Department of Thoracic Surgery, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People's Hospital, Yancheng 224000, P.R. China
- Medical School of Nantong University, Nantong, 226007, P.R. China
| | - Xia Li
- Department of General Medicine, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People's Hospital, Yancheng 224000, P.R. China
| | - Jianxiang Song
- Department of Thoracic Surgery, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People's Hospital, Yancheng 224000, P.R. China
| |
Collapse
|