51
|
Sheikh M, Saiyyad A, Aliunui A, Jirvankar PS. The evolving landscape of oncolytic virus immunotherapy: combinatorial strategies and novel engineering approaches. Med Oncol 2025; 42:190. [PMID: 40314865 DOI: 10.1007/s12032-025-02746-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Accepted: 04/25/2025] [Indexed: 05/03/2025]
Abstract
Oncolytic viruses (OVs) are a promising class of cancer therapy, exploiting their abilities to selectively infect and kill cancer cells while stimulating antitumor immune responses. The current assessment explores the changing horizons of OV immunotherapy, focusing on recent advances in technology plans to improve OV projects and combined approaches to improve curative efficacy. We discuss how OVs induce direct oncolysis and promote the release of tumor-associated antigens, leading to the activation of both innate and adaptive immunity. Special attention shall be given to programs for arm OVs to express curative genes, modify the tumor microenvironment and overcome immunosuppression. Moreover, we assess the synergies of uniting OVs with other immunotherapeutic techniques, such as immune checkpoint inhibitors and cell therapy, to improve tolerant outcomes. The present assessment provides an understanding of the relevant declaration of the OV analysis, highlighting the main obstacles and the future directions for the development of other capable and targeted cancer immunotherapy.
Collapse
Affiliation(s)
- Mujibullah Sheikh
- Datta Meghe College of Pharmacy DMIHER (Deemed to be University), Wardha, Maharashtra, 442001, India.
| | - Arshiya Saiyyad
- Datta Meghe College of Pharmacy DMIHER (Deemed to be University), Wardha, Maharashtra, 442001, India
| | - Aimé Aliunui
- Datta Meghe College of Pharmacy DMIHER (Deemed to be University), Wardha, Maharashtra, 442001, India
| | - Pranita S Jirvankar
- Datta Meghe College of Pharmacy DMIHER (Deemed to be University), Wardha, Maharashtra, 442001, India
| |
Collapse
|
52
|
Lorenc P, Dams-Kozlowska H, Guzniczak N, Florczak-Substyk A. Application of nanoparticles to target tumor blood vessels as a promising cancer treatment strategy. Biomed Pharmacother 2025; 186:118038. [PMID: 40215646 DOI: 10.1016/j.biopha.2025.118038] [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: 02/10/2025] [Revised: 03/31/2025] [Accepted: 04/03/2025] [Indexed: 04/25/2025] Open
Abstract
Cancer remains one of the leading causes of death worldwide and poses a significant challenge to effective treatment due to its complexity. Angiogenesis, the formation of new blood vessels, is a critical process in tumor growth and metastasis. The VEGF/VEGFR pathway plays a crucial role in regulating angiogenesis. Many anti-angiogenesis agents, including monoclonal antibodies and tyrosine kinase inhibitors, have been investigated for the treatment of various cancers. However, they face significant limitations such as limited bioavailability and drug resistance. Nanoparticles have emerged as a promising tool for effective drug delivery while minimizing systemic side effects. This review explores the application of nanoparticles dedicated to angiogenesis-targeted cancer therapy, particularly targeting the VEGF/VEGFR pathway. We describe drug delivery systems based on inorganic, lipid, and polymeric nanoparticles. Moreover, special attention is given to functionalized nanoparticles, which can precisely target numerous proteins that are significantly overexpressed on the surfaces of endothelial cells, tumors, or other cells in the tumor microenvironment. We summarize a series of nanoparticles designed for selective targeting of tumor vasculature, emphasizing the challenges faced by anti-angiogenic cancer therapies.
Collapse
Affiliation(s)
- Patryk Lorenc
- Chair of Medical Biotechnology, Department of Cancer Immunology, Poznan University of Medical Sciences, 8 Rokietnicka St, Poznan 60-806, Poland; Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St, Poznan 61-866, Poland; Doctoral School, Poznan University of Medical Sciences, 70 Bukowska St, Poznan 60-812, Poland
| | - Hanna Dams-Kozlowska
- Chair of Medical Biotechnology, Department of Cancer Immunology, Poznan University of Medical Sciences, 8 Rokietnicka St, Poznan 60-806, Poland; Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St, Poznan 61-866, Poland
| | - Natalia Guzniczak
- Chair of Medical Biotechnology, Department of Cancer Immunology, Poznan University of Medical Sciences, 8 Rokietnicka St, Poznan 60-806, Poland
| | - Anna Florczak-Substyk
- Chair of Medical Biotechnology, Department of Cancer Immunology, Poznan University of Medical Sciences, 8 Rokietnicka St, Poznan 60-806, Poland; Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St, Poznan 61-866, Poland.
| |
Collapse
|
53
|
Janes KA, Lazzara MJ. Systems Biology of the Cancer Cell. Annu Rev Biomed Eng 2025; 27:1-28. [PMID: 39689262 DOI: 10.1146/annurev-bioeng-103122-030552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
Questions in cancer have engaged systems biologists for decades. During that time, the quantity of molecular data has exploded, but the need for abstractions, formal models, and simplifying insights has remained the same. This review brings together classic breakthroughs and recent findings in the field of cancer systems biology, focusing on cancer cell pathways for tumorigenesis and therapeutic response. Cancer cells mutate and transduce information from their environment to alter gene expression, metabolism, and phenotypic states. Understanding the molecular architectures that make each of these steps possible is a long-term goal of cancer systems biology pursued by iterating between quantitative models and experiments. We argue that such iteration is the best path to deploying targeted therapies intelligently so that each patient receives the maximum benefit for their cancer.
Collapse
Affiliation(s)
- Kevin A Janes
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia, USA
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA; ,
| | - Matthew J Lazzara
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia, USA
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA; ,
| |
Collapse
|
54
|
Vinogradov AE, Anatskaya OV. "Cell dedifferentiation" versus "evolutionary reversal" theories of cancer: The direct contest of transcriptomic features. Int J Cancer 2025; 156:1802-1813. [PMID: 39888036 DOI: 10.1002/ijc.35352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 01/07/2025] [Accepted: 01/20/2025] [Indexed: 02/01/2025]
Abstract
Cell dedifferentiation is considered an important hallmark of cancer. The atavistic reversal to a unicellular-like (UC) state is a less widely accepted concept, so far not included in the conventional hallmarks. The activated expression of ontogenetically earlier and evolutionary earlier genes in cancers supports both theories because ontogenesis partially recapitulates phylogenesis during cell differentiation (the cellular biogenetic law). We directly contested both types of gene signatures in stem vs. differentiated and cancer vs. normal cells, using meta-analysis of human single-cell transcriptomes (totally, 38 pairwise comparisons involving over 18,600 cells). Because compared cells can differ in proliferation rate, the correction for cell cycle activity was applied. Taken together as multiple variables in stem vs. differentiated cells analyses, the ontogenetic signature excluded the UC signature from predictive variables, usually even forcing it to change the sign of prediction (from plus to minus). In contrast, in cancer vs. normal cells, the UC signature excluded the ontogenetic signature. Thus, the direct contest decided in favor of the atavistic theory and placed a UC-like state as a central hallmark of cancer, which has a plausible evolutionarily formed mechanism (UC attractor) and can generate other hallmarks. These data suggest a paradigm shift in the understanding of oncogenesis and propose an integrative framework for cancer research. In a practical sense, the upregulation of UC signature over ontogenetic signature indicates potential tumorigenicity, which can be used in early diagnostics and regenerative medicine. For therapy, these results suggest the UC center of cellular networks as a universal target.
Collapse
Affiliation(s)
| | - Olga V Anatskaya
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russia
| |
Collapse
|
55
|
Zhang K, Zhao T, Riaz F, Li Y, Wei P, Fang X, Zhou Z, Kou W, Pan F. Neuritin-specific antibody impedes the Treg-mediated suppression of anti-tumor immunity and enhances response to anti-PD1. Mol Immunol 2025; 181:148-159. [PMID: 40153952 DOI: 10.1016/j.molimm.2025.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 03/08/2025] [Accepted: 03/19/2025] [Indexed: 04/01/2025]
Abstract
Regulatory T cells (Tregs) and effector T cells play critical roles in tumor immunity, with Tregs suppressing immune responses and contributing to an immunosuppressive tumor microenvironment (TME). Neuritin-1 (Nrn), a neuropeptide, has been identified to enhance Treg expansion. However, its role in T cell biology and tumor development remains unclear. We demonstrated that Nrn is highly expressed in the in-vitro-induced Tregs (iTregs). Functionally, Nrn promoted iTreg differentiation in a dose-dependent manner, while Nrn deletion or anti-Nrn antibody treatment significantly inhibited iTreg differentiation. Additionally, Nrn suppressed IL-2 transcription and secretion in T cells, impairing T cell activation and pro-inflammatory cytokine production. Treg-specific Nrn knockout mice exhibited reduced B16 melanoma tumor growth, decreased Treg infiltration, and increased effector T cell infiltration. Conversely, overexpression of Nrn accelerated B16 melanoma tumor progression by enhancing Treg-mediated suppression. Importantly, we developed the first anti-Nrn antibody, which effectively reduced tumour growth, decreased Treg infiltration, and enhanced effector T-cell activity. Importantly, anti-Nrn synergistically worked with anti-PD1 and improved the anti-PD1 response by reducing Tregs and increasing effector function in tumor-infiltrated T cells, resulting in enhanced tumor regression. Our findings identify Nrn as a critical regulator of Treg differentiation and effector T cell suppression, contributing to tumor progression. Targeting Nrn alone or combined with anti-PD1 therapy represents a promising strategy to enhance anti-tumor immunity.
Collapse
Affiliation(s)
- Kaimin Zhang
- Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), 1068 Xueyuan Avenue, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Taowen Zhao
- Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), 1068 Xueyuan Avenue, Shenzhen 518055, China
| | - Fraooq Riaz
- Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology (SUAT), China
| | - Yikui Li
- Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), 1068 Xueyuan Avenue, Shenzhen 518055, China
| | - Ping Wei
- Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), 1068 Xueyuan Avenue, Shenzhen 518055, China; Department of Pediatric Otolaryngology Head and Neck Surgery, West China Second University Hospital, Sichuan University, 1416, Section 1, Chenglong Avenue, Chengdu 610066, China
| | - Xiang Fang
- Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), 1068 Xueyuan Avenue, Shenzhen 518055, China
| | - Zhiyi Zhou
- Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), 1068 Xueyuan Avenue, Shenzhen 518055, China
| | - Wei Kou
- Department of Pediatric Otolaryngology Head and Neck Surgery, West China Second University Hospital, Sichuan University, 1416, Section 1, Chenglong Avenue, Chengdu 610066, China.
| | - Fan Pan
- Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), 1068 Xueyuan Avenue, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China; Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology (SUAT), China.
| |
Collapse
|
56
|
Klümper N, Cox A, Sjödahl G, Roghmann F, Bolenz C, Hartmann A, Grünwald V, Faltas BM, Hölzel M, Eckstein M. Pre-treatment metastatic biopsy: a step towards precision oncology for urothelial cancer. Nat Rev Urol 2025; 22:256-267. [PMID: 39472646 DOI: 10.1038/s41585-024-00951-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2024] [Indexed: 05/10/2025]
Abstract
Early metastatic spread and clonal expansion of individual mutations result in a heterogeneous tumour landscape in metastatic urothelial cancer (mUC). Substantial molecular heterogeneity of common drug targets, such as membranous NECTIN4, FGFR3 mutations, PDL1 or immune phenotypes, has been documented between primary and metastatic tumours. However, translational and clinical studies frequently do not account for such heterogeneity and often investigate primary tumour samples that might not be representative in patients with mUC. We propose this as a potential factor for why many biomarkers for mUC have failed to be integrated into clinical practice. Fresh pre-treatment metastatic biopsies enable the capturing of prevailing tumour biology in real time. The characterization of metastatic tumour samples can improve response prediction to immunotherapy, the anti-NECTIN4 antibody-drug conjugate enfortumab vedotin and the FGFR inhibitor erdafitinib. Routine metastatic biopsy can thus improve the precision of identifying driver druggable alterations, thus improving treatment selection for patients with mUC.
Collapse
Affiliation(s)
- Niklas Klümper
- Department of Urology and Pediatric Urology, University Hospital Bonn, Bonn, Germany.
- Institute of Experimental Oncology, University Hospital Bonn, Bonn, Germany.
| | - Alexander Cox
- Department of Urology and Pediatric Urology, University Hospital Bonn, Bonn, Germany
| | - Gottfrid Sjödahl
- Department of Translational Medicine, Division of Urological Research, Lund University, Lund, Sweden
| | - Florian Roghmann
- Department of Urology, Marien Hospital, Ruhr-University Bochum, Herne, Germany
| | - Christian Bolenz
- Department of Urology and Paediatric Urology, University Hospital Ulm, University of Ulm, Ulm, Germany
| | - Arndt Hartmann
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Viktor Grünwald
- Clinic for Internal Medicine (Tumour Research) and Clinic for Urology, Interdisciplinary Genitourinary Oncology at the West-German Cancer Center, Essen University Hospital, Essen, Germany
| | - Bishoy M Faltas
- Department of Hematology/Oncology, Weill-Cornell Medicine, New York, NY, USA
| | - Michael Hölzel
- Institute of Experimental Oncology, University Hospital Bonn, Bonn, Germany
| | - Markus Eckstein
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
57
|
Yoshida K, Yokoi A, Suzuki H, Tamauchi S, Kitagawa M, Inami E, Nakayama J, Mori Y, Okamoto K, Suzuki Y, Yoshida H, Kato T, Kajiyama H, Yamamoto Y. Single-Nucleus RNA Sequencing and Spatial Transcriptomics for Squamous Cell Carcinoma Arising From Ovarian Mature Teratoma. Cancer Sci 2025; 116:1339-1351. [PMID: 39948701 PMCID: PMC12044660 DOI: 10.1111/cas.70022] [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: 11/29/2024] [Revised: 02/01/2025] [Accepted: 02/05/2025] [Indexed: 05/02/2025] Open
Abstract
Squamous cell carcinoma arising from mature teratoma (SCC-MT) is a rare ovarian malignancy. The detailed molecular pathology of SCC-MT is not well understood. Moreover, the prognosis of the patients remains poor because no standard treatment has been established. In this study, we performed single-nucleus RNA sequencing and spatial transcriptomics using clinical SCC-MT samples to identify novel therapeutic candidates. snRNA-seq revealed three epithelial cell clusters, of which one was significantly associated with epidermis and keratinocyte development. Moreover, spatial transcriptomics revealed that the epithelial-mesenchymal transition was significantly inhibited, and the MYC and E2F targets were significantly activated in cancer spots on specimen sections. We focused on KLF5, which was one of the upregulated genes in cancer cells, and performed a functional analysis using NOSCC-1, a cell line derived from an SCC-MT. KLF5 downregulation significantly decreased cell proliferation and increased apoptosis. Furthermore, we previously identified miR-145-5p as a downregulated miRNA in SCC-MT. We demonstrated that miR-145-5p overexpression attenuated cell proliferation and decreased KLF5 expression. In conclusion, through multi-omics analyses, we identified unique gene expression profiles of SCC-MT and determined a role for KLF5 in SCC-MT development. Therefore, KLF5-related factors may be novel therapeutic targets, and further studies are needed to improve the diagnosis and treatment of SCC-MT.
Collapse
Affiliation(s)
- Kosuke Yoshida
- Department of Obstetrics and GynecologyNagoya University Graduate School of MedicineNagoyaJapan
- Institute for Advanced ResearchNagoya UniversityNagoyaJapan
- Laboratory of Integrative OncologyNational Cancer Center Research InstituteTokyoJapan
| | - Akira Yokoi
- Department of Obstetrics and GynecologyNagoya University Graduate School of MedicineNagoyaJapan
- Institute for Advanced ResearchNagoya UniversityNagoyaJapan
| | - Hironori Suzuki
- Department of Obstetrics and GynecologyNagoya University Graduate School of MedicineNagoyaJapan
- Laboratory of Integrative OncologyNational Cancer Center Research InstituteTokyoJapan
| | - Satoshi Tamauchi
- Department of Obstetrics and GynecologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Masami Kitagawa
- Department of Obstetrics and GynecologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Eri Inami
- Department of Obstetrics and GynecologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Jun Nakayama
- Laboratory of Integrative OncologyNational Cancer Center Research InstituteTokyoJapan
- Department of Oncogenesis and Growth RegulationResearch Institute, Osaka International Cancer InstituteOsakaJapan
| | - Yutaro Mori
- Advanced Comprehensive Research OrganizationTeikyo UniversityTokyoJapan
| | - Koji Okamoto
- Advanced Comprehensive Research OrganizationTeikyo UniversityTokyoJapan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier SciencesThe University of TokyoJapan
| | - Hiroshi Yoshida
- Department of Diagnostic PathologyNational Cancer Center HospitalTokyoJapan
| | - Tomoyasu Kato
- Department of GynecologyNational Cancer Center HospitalTokyoJapan
| | - Hiroaki Kajiyama
- Department of Obstetrics and GynecologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Yusuke Yamamoto
- Laboratory of Integrative OncologyNational Cancer Center Research InstituteTokyoJapan
| |
Collapse
|
58
|
Lu X, Shui KM, Ji H, Chen JY, Yan C, Li P, Xu T. Nerve infiltration of bladder cancer predicts response to immunotherapy. Biochem Biophys Res Commun 2025; 761:151687. [PMID: 40179739 DOI: 10.1016/j.bbrc.2025.151687] [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: 03/15/2025] [Accepted: 03/22/2025] [Indexed: 04/05/2025]
Abstract
Immune checkpoint therapy (ICT), particularly Programmed Death-1 (PD1) antibody treatment, has revolutionized the clinical management of many previously undruggable types bladder cancer (BC). However, it remains unclear whether nerve-invaded BC patients benefit from immunotherapy. Here, we utilized public databases to show that BC patients with high neural signal expression are associated with more advanced tumor stages. Analysis of patient tumor samples revealed significant neural expression within classical tertiary lymphoid structures, and this was inversely correlated with the immune levels. In a denervated mouse bladder tumor model, we found that nerve ablation significantly inhibited tumor progression. Single-nucleus RNA sequencing data revealed that nerve fibers suppressed the activity of immune cells within the tumor microenvironment, while denervation alleviated immune suppression and restored cytotoxic activity. Denervation enhanced the immune response by downregulating Pdcd1 (encoded Pd1) expression in natural killer (NK) cells. Pathological analysis of patient tumor samples further confirmed that higher neural abundance was strongly correlated with PD1 expression. Finally, through database analysis and clinical sample validation, we demonstrated that BC patients with high neural signaling showed better responses to ICT. Overall, this study highlights the potential for targeted immunotherapy in nerve-invasive BC, providing a novel framework for enhancing the effectiveness of ICT in treating this subset of bladder cancer.
Collapse
Affiliation(s)
- Xinyi Lu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center, ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, China.
| | - Kun-Ming Shui
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center, ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, China
| | - Hao Ji
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jia-Yu Chen
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center, ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, China
| | - Chao Yan
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center, ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, China.
| | - Pengchao Li
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China; Jiangsu Province (Suqian) Hospital, Suqian, Jiangsu, China.
| | - Tao Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China.
| |
Collapse
|
59
|
Sharma R, Chirom O, Mujib A, Prasad M, Prasad A. UFMylation: Exploring a lesser known post translational modification. PLANT SCIENCE : AN INTERNATIONAL JOURNAL OF EXPERIMENTAL PLANT BIOLOGY 2025; 354:112435. [PMID: 39993644 DOI: 10.1016/j.plantsci.2025.112435] [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: 11/13/2024] [Revised: 02/06/2025] [Accepted: 02/18/2025] [Indexed: 02/26/2025]
Abstract
Ubiquitination is a highly conserved post-translational modification (PTM) in which ubiquitin (Ub) is covalently attached to substrate proteins resulting in the alteration of protein structure, function, and stability. Another class of PTM mediated by ubiquitin-like proteins (UBLs) has gained significant attention among researchers in recent years. This article focuses on one such UBL-mediated PTM i.e. UFMylation. The enzymatic mechanism of UFMylation is similar to ubiquitination, involving three steps regulated by three different enzymes. In plants, reports suggest that UFMylation is predominantly involved in maintaining ER homeostasis including ER-phagy. However, studies related to this PTM are limited and future studies might reveal other molecular pathways regulated by UFMylation.
Collapse
Affiliation(s)
- Rohit Sharma
- Department of Botany, Kurukshetra University, Kurukshetra, India
| | - Oceania Chirom
- Department of Botany, Jamia Hamdard University, New Delhi, India
| | - Abdul Mujib
- Department of Botany, Jamia Hamdard University, New Delhi, India
| | - Manoj Prasad
- Department of Genetics, University of Delhi South Campus, New Delhi, India; National Institute of Plant Genome Research, New Delhi, India.
| | - Ashish Prasad
- Department of Botany, Kurukshetra University, Kurukshetra, India.
| |
Collapse
|
60
|
Mou D, Wu S, Chen Y, Wang Y, Dai Y, Tang M, Teng X, Bai S, Bai X. Roles of PEG10 in cancer and neurodegenerative disorder (Review). Oncol Rep 2025; 53:60. [PMID: 40183369 PMCID: PMC11976372 DOI: 10.3892/or.2025.8893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 03/06/2025] [Indexed: 04/05/2025] Open
Abstract
Paternally expressed gene 10 (PEG10) is an imprinting gene. In addition to its known roles in placental development, as well as mouse embryonic stem cell and trophoblast stem cell differentiation, PEG10 has recently been shown to have significance in cancers. High expression of PEG10 is observed in various cancer types and is associated with poor prognosis. Of note, disruption of PEG10 expression leads to increased apoptosis, as well as decreased proliferation, invasion and migration of cancer cells. PEG10 is expected to become a target for cancer and neurodegenerative disorder therapy. This article reviewed the latest progress in the role of PEG10 in cancers.
Collapse
Affiliation(s)
- Dachao Mou
- Laboratory of Human Disease and Immunotherapies, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Institute of Inflammation and Immunology (I), Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Shasha Wu
- Laboratory of Human Disease and Immunotherapies, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Institute of Inflammation and Immunology (I), Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yanqiong Chen
- Laboratory of Human Disease and Immunotherapies, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Institute of Inflammation and Immunology (I), Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yun Wang
- Laboratory of Human Disease and Immunotherapies, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Institute of Inflammation and Immunology (I), Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yufang Dai
- Laboratory of Human Disease and Immunotherapies, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Institute of Inflammation and Immunology (I), Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Min Tang
- Division of Thoracic Tumor Multimodality Treatment and Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xiu Teng
- Laboratory of Human Disease and Immunotherapies, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Institute of Inflammation and Immunology (I), Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Shijun Bai
- Department of Agriculture Forestry and Food Engineering, Yibin University, Lingang Economic and Technological Development Zone, Yibin, Sichuan 644000, P.R. China
| | - Xiufeng Bai
- Laboratory of Human Disease and Immunotherapies, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Institute of Inflammation and Immunology (I), Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
61
|
Martins Rodrigues F, Terekhanova NV, Imbach KJ, Clauser KR, Esai Selvan M, Mendizabal I, Geffen Y, Akiyama Y, Maynard M, Yaron TM, Li Y, Cao S, Storrs EP, Gonda OS, Gaite-Reguero A, Govindan A, Kawaler EA, Wyczalkowski MA, Klein RJ, Turhan B, Krug K, Mani DR, Leprevost FDV, Nesvizhskii AI, Carr SA, Fenyö D, Gillette MA, Colaprico A, Iavarone A, Robles AI, Huang KL, Kumar-Sinha C, Aguet F, Lazar AJ, Cantley LC, Marigorta UM, Gümüş ZH, Bailey MH, Getz G, Porta-Pardo E, Ding L. Precision proteogenomics reveals pan-cancer impact of germline variants. Cell 2025; 188:2312-2335.e26. [PMID: 40233739 DOI: 10.1016/j.cell.2025.03.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 04/29/2024] [Accepted: 03/13/2025] [Indexed: 04/17/2025]
Abstract
We investigate the impact of germline variants on cancer patients' proteomes, encompassing 1,064 individuals across 10 cancer types. We introduced an approach, "precision peptidomics," mapping 337,469 coding germline variants onto peptides from patients' mass spectrometry data, revealing their potential impact on post-translational modifications, protein stability, allele-specific expression, and protein structure by leveraging the relevant protein databases. We identified rare pathogenic and common germline variants in cancer genes potentially affecting proteomic features, including variants altering protein abundance and structure and variants in kinases (ERBB2 and MAP2K2) impacting phosphorylation. Precision peptidome analysis predicted destabilizing events in signal-regulatory protein alpha (SIRPA) and glial fibrillary acid protein (GFAP), relevant to immunomodulation and glioblastoma diagnostics, respectively. Genome-wide association studies identified quantitative trait loci for gene expression and protein levels, spanning millions of SNPs and thousands of proteins. Polygenic risk scores correlated with distal effects from risk variants. Our findings emphasize the contribution of germline genetics to cancer heterogeneity and high-throughput precision peptidomics.
Collapse
Affiliation(s)
- Fernanda Martins Rodrigues
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Nadezhda V Terekhanova
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Kathleen J Imbach
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain; Universitat Autonoma de Barcelona, Barcelona, Spain
| | | | - Myvizhi Esai Selvan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Thoracic Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Isabel Mendizabal
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain; Translational Prostate Cancer Research Lab, CIC bioGUNE-Basurto, Biocruces Bizkaia Health Research Institute, Derio, Spain
| | - Yifat Geffen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Cancer Center and Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Yo Akiyama
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Tomer M Yaron
- Meyer Cancer Center, Department of Medicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Yize Li
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Song Cao
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Erik P Storrs
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Olivia S Gonda
- Department of Biology, Brigham Young University, Salt Lake City, UT, USA
| | - Adrian Gaite-Reguero
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain
| | - Akshay Govindan
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Emily A Kawaler
- Applied Bioinformatics Laboratories, New York University Langone Health, New York City, NY, USA
| | - Matthew A Wyczalkowski
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Robert J Klein
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Berk Turhan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Karsten Krug
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - D R Mani
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Alexey I Nesvizhskii
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA; Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Steven A Carr
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - David Fenyö
- Institute for Systems Genetics, NYU Grossman School of Medicine, New York, NY, USA
| | | | - Antonio Colaprico
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL, USA; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Antonio Iavarone
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Neurological Surgery, Department of Biochemistry and Molecular Biology, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Ana I Robles
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, Rockville, MD, USA
| | - Kuan-Lin Huang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Transformative Disease Modeling, Tisch Cancer Institute, Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chandan Kumar-Sinha
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA; Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | | | - Alexander J Lazar
- Departments of Pathology and Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Urko M Marigorta
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Zeynep H Gümüş
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Thoracic Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Matthew H Bailey
- Department of Biology, Brigham Young University, Salt Lake City, UT, USA.
| | - Gad Getz
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Cancer Center and Department of Pathology, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| | - Eduard Porta-Pardo
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain; Barcelona Supercomputing Center (BSC), Barcelona, Spain.
| | - Li Ding
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA; Siteman Cancer Center, Washington University in St. Louis, Saint Louis, MO, USA.
| |
Collapse
|
62
|
Lu X, Friedrich LJ, Efferth T. Natural products targeting tumour angiogenesis. Br J Pharmacol 2025; 182:2094-2136. [PMID: 37680009 DOI: 10.1111/bph.16232] [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: 06/23/2023] [Revised: 08/15/2023] [Accepted: 08/28/2023] [Indexed: 09/09/2023] Open
Abstract
Tumour angiogenesis is the formation of new blood vessels to support the growth of a tumour. This process is critical for tumour progression and metastasis, making it an attractive approach to cancer therapy. Natural products derived from plants, animals or microorganisms exert anti-angiogenic properties and can be used to inhibit tumour growth and progression. In this review, we comprehensively report on the current status of natural products against tumour angiogenesis from four perspectives until March 2023: (1) the role of pro-angiogenic factors and antiangiogenic factors in tumour angiogenesis; (2) the development of anti-tumour angiogenesis therapy (monoclonal antibodies, VEGFR-targeted small molecules and fusion proteins); (3) the summary of anti-angiogenic natural agents, including polyphenols, polysaccharides, alkaloids, terpenoids, saponins and their mechanisms of action, and (4) the future perspectives of anti-angiogenic natural products (bioavailability improvement, testing of dosage and side effects, combination use and discovery of unique natural-based compounds). Our review aims to better understand the potential of natural products for drug development in inhibiting tumour angiogenesis and further aid the effective transition of these outcomes into clinical trials. LINKED ARTICLES: This article is part of a themed issue Natural Products and Cancer: From Drug Discovery to Prevention and Therapy. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v182.10/issuetoc.
Collapse
Affiliation(s)
- Xiaohua Lu
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Lara Johanna Friedrich
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
63
|
Mojumder A, Shahrear S, Md. Khademul Islam ABM. Deciphering the Role of Putative Novel miRNAs Encoded From the Newly Found Genomic Regions of T2T-CHM13 in the Progression of Collecting Duct Renal Cell Carcinoma. Cancer Med 2025; 14:e70925. [PMID: 40304310 PMCID: PMC12042112 DOI: 10.1002/cam4.70925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/19/2025] [Accepted: 04/21/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND Collecting duct renal cell carcinoma (cdRCC) is a rare and aggressive renal cancer subtype. The molecular mechanisms underlying cdRCC remain poorly understood, which presents a significant challenge for the development of effective treatment strategies. Advances in genome sequencing, particularly the discovery of new genomic regions in the T2T-CHM13 reference genome, have provided an opportunity to expand our understanding of this disease. AIMS Our study specifically aims to investigate the role of miRNAs encoded by these regions, proposing them as potential epigenetic regulators in the pathogenesis of cdRCC. METHODS We used bioinformatic pipelines and small RNA-seq data analysis to predict novel miRNAs from the newly discovered genomic regions of T2T-CHM13. RNA-seq analysis of cdRCC tumors was performed to identify differentially expressed genes, and target prediction tools were used to find miRNA-mRNA interactions. Functional enrichment analyses were conducted to characterize the biological pathways affected. RESULTS AND DISCUSSION Using computational approaches, we predicted 156 novel miRNAs from T2T-CHM13's newly resolved sequences. RNA-seq and miRNA-mRNA interaction analyses identified 345 downregulated genes targeted by novel miRNAs and 395 downregulated genes targeted by known miRNAs. A comprehensive functional enrichment analysis of these perturbed genes revealed distinctive pathways, including cGMP-PKG signaling, calcium signaling, adipocytokine signaling, PPAR signaling, and apelin signaling, all of which are implicated in tumorigenesis. Furthermore, Gene Ontology analysis linked miRNA-targeted genes to disrupted cell-cell junctions and adhesion, providing a mechanistic explanation for aggressive invasion and metastasis in cdRCC. Additionally, a significant number of the target genes involved in metabolic and ion transport pathways were perturbed, explaining metabolic alterations in the cancer cells. We also identified 15 tumor suppressor genes downregulated by novel miRNAs, 6 of which were uniquely targeted, highlighting the potential of these miRNAs as cdRCC-specific biomarkers. CONCLUSION In conclusion, our study offers valuable insights into cdRCC biology from an epigenetic perspective, laying the groundwork for future research aimed at developing targeted therapies.
Collapse
Affiliation(s)
- Anik Mojumder
- Department of Genetic Engineering and BiotechnologyUniversity of DhakaDhakaBangladesh
| | - Sazzad Shahrear
- Department of Genetic Engineering and BiotechnologyUniversity of DhakaDhakaBangladesh
| | | |
Collapse
|
64
|
Zhang C, Zhou W, Xu H, Xu J, Li J, Liu X, Lu X, Dai J, Jiang Y, Wang W, Zhang E, Guo R. Cancer-associated fibroblasts promote EGFR-TKI resistance via the CTHRC1/glycolysis/H3K18la positive feedback loop. Oncogene 2025; 44:1400-1414. [PMID: 40011576 PMCID: PMC12052591 DOI: 10.1038/s41388-025-03318-y] [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: 09/22/2024] [Revised: 01/20/2025] [Accepted: 02/18/2025] [Indexed: 02/28/2025]
Abstract
Acquired resistance to epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) remains a major challenge in the treatment of lung cancer. Cancer associated fibroblasts (CAFs) play a key role in promoting resistance to anti-cancer therapies. This study identified a subpopulation of CAFs characterized by the overexpression of collagen triple helix repeat-containing 1 (CTHRC1) through single-cell RNA sequencing of lung cancer patients undergoing EGFR-TKI treatment. These CTHRC1+ CAFs were enriched in drug-resistant tumors. Mechanistically, CTHRC1+ CAFs enhance the glycolytic activity of cancer cells by activating the TGF-β/Smad3 signaling pathway. Excess lactate produced in the process of glycolysis further upregulates CTHRC1 expression in CAFs through histone lactylation, creating a positive feedback loop that sustains EGFR-TKI resistance. The study also demonstrated that Gambogenic Acid, a natural compound, can disrupt this feedback loop, thereby improving the efficacy of EGFR-TKI therapy. Additionally, the presence of CTHRC1+ CAFs in tumor tissues could serve as a biomarker for predicting the response to EGFR-TKI therapy and patient prognosis. Overall, this study highlights the significant role of CAFs in EGFR-TKI resistance and suggests that targeting CTHRC1+ CAFs could be a promising strategy to overcome drug resistance in lung cancer.
Collapse
Affiliation(s)
- Chen Zhang
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Wenxin Zhou
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Hai Xu
- Department of Radiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Jiali Xu
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Jun Li
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xinyin Liu
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xiyi Lu
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Jiali Dai
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yuqin Jiang
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Wei Wang
- Department of Thoracic Surgery, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Erbao Zhang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China.
| | - Renhua Guo
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
65
|
Huang B, Shen W, Jia Y, Qin L, Wang H, Sun Q, Xiao Z, Zhang R, Wang H. LDHAα, a lactate dehydrogenase A isoform, promotes glycolysis and tumor progression. FEBS J 2025; 292:2223-2236. [PMID: 39828959 DOI: 10.1111/febs.17374] [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: 05/24/2024] [Revised: 09/25/2024] [Accepted: 12/16/2024] [Indexed: 01/22/2025]
Abstract
Lactate dehydrogenase A (LDHA) is upregulated in multiple cancer types and contributes to the Warburg effect. Several studies have found that many tumor-related genes have subtypes and play important roles in promoting cancer development. Here, we identified a novel LDHA transcript, which produced a new protein 3 kDa larger than LDHA, which we named LDHAα. We found that multiple cancer cell lines express LDHAα, and ectopic expression of LDHAα led to a higher proliferation and migration rate in vitro. Ectopic expression of LDHAα could also promote tumor cell growth in vivo. Conversely, deletion of LDHAα by CRISPR-sgRNA significantly inhibited the growth of tumor cells. LDHAα was found to be mainly located in the cytoplasm, and overexpression or deletion of LDHAα could significantly affect the glucose uptake and lactate production of tumor cells. Further investigation showed that c-MYC and FOXM1 could markedly modulate the expression of both LDHA and LDHAα, especially c-MYC. We found that a small molecular compound targeting LDHA could also inhibit the enzyme activity of LDHAα. LDHAα, LDHA and c-MYC expression was significantly higher in human acute lymphocytic leukemia and colorectal cancer tissue specimens compared to normal controls. In conclusion, our study identified LDHAα as a subtype of LDHA and highlighted its critical role in tumor metabolism, providing a potential new therapeutic target for tumor diagnosis and treatment.
Collapse
Affiliation(s)
- Bingqing Huang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Wencui Shen
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Hospital & Eye Institute, NanKai University, Tianjin, China
| | - Yujiao Jia
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Li Qin
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Haoxu Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Qi Sun
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Zhijian Xiao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Rongxin Zhang
- Laboratory of Immunology and Inflammation, Department of Biotechnology, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Huijun Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| |
Collapse
|
66
|
Liu M, Li TZ, Xu C. The role of tumor-associated fibroblast-derived exosomes in chemotherapy resistance of colorectal cancer and its application prospect. Biochim Biophys Acta Gen Subj 2025; 1869:130796. [PMID: 40122307 DOI: 10.1016/j.bbagen.2025.130796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/03/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
Colorectal cancer (CRC) is the second most common malignant tumor in the world. With its increasing incidence and younger age trend, its impact on human health has been paid more and more attention. Currently, we have a variety of chemotherapy drugs that can be used to treat colorectal cancer. However, the drug resistance of colorectal cancer has become a significant factor affecting its cure rate. Some studies have reported that exosomes are related to the occurrence of drug resistance. However, the exact mechanism is not precise. Therefore, we focused on the role of cancer associated-fibroblast-derived (CAFs-derived) exosomes in colorectal progression. It was found that cancer cells transmit information through exosome interaction and induce chemotherapy resistance by promoting epithelial-mesenchymal transition (EMT), up-regulating the Wnt/β-catenin signaling pathway, transforming growth factor-β1 (TGF-β1) pathway, promoting angiogenesis and other possible molecular mechanisms. In addition, in terms of clinical significance and therapeutic strategies, we explore the clinical relevance of CAFs-derived exosomes in colorectal cancer patients and their potential as potential biomarkers for predicting chemotherapy response. We also provide a new possible direction for overcoming chemotherapy resistance in colorectal cancer by targeting CAFs-derived exosomes.
Collapse
Affiliation(s)
- Meichen Liu
- The Second Clinical Medical College, Nanchang University, NanChang, China
| | - Teng-Zheng Li
- Department of Gastroenterology, The second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, NanChang, China
| | - Congcong Xu
- Department of Cardiovascular Medicine, The second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, NanChang, China.
| |
Collapse
|
67
|
Schulze K, Rose TD, Adlung L, Peschka M, Pagani F, Gorgulho J, Fründt TW, Labgaa I, Haber PK, Zimpel C, Castven D, Weinmann A, Garzia-Lezana T, Waldmann M, Renné T, Voß H, Moritz M, Orlikowski D, Schlüter H, Baumbach J, Schwartz M, Lohse AW, Huber S, Sangro B, Macias RI, Izquierdo-Sanchez L, Banales JM, Wege H, Marquardt JU, Villanueva A, Pauling JK, von Felden J. Metabolomic liquid biopsy dynamics predict early-stage HCC and actionable candidates of human hepatocarcinogenesis. JHEP Rep 2025; 7:101340. [PMID: 40290517 PMCID: PMC12023797 DOI: 10.1016/j.jhepr.2025.101340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/16/2025] [Accepted: 01/22/2025] [Indexed: 04/30/2025] Open
Abstract
Background & Aims Actionable candidates of hepatocarcinogenesis remain elusive, and tools for early detection are suboptimal. Our aim was to demonstrate that serum metabolome profiles reflect the initiation of hepatocellular carcinoma (HCC) and enable the identification of biomarkers for early HCC detection and actionable candidates for chemoprevention. Methods This global cohort study included 654 patients and 801 biospecimens. Following serum metabolome profiling across the spectrum of hepatocarcinogenesis, we conducted a phase II biomarker case-control study for early HCC detection. Findings were independently validated through in silico analysis, mRNA sequencing, and proteome profiling of primary HCC and non-tumoral tissue, and in vitro experiments. Results Aspartic acid, glutamic acid, taurine, and hypoxanthine were differentially abundant in the serum across chronic liver disease, cirrhosis, initial HCC, and progressed HCC, independent of sex, age, and etiology. In a phase II biomarker case-control study, a blood-based metabolite signature yielded an AUC of 94% to discriminate between patients with early-stage HCC and controls with cirrhosis, including independent validation. Unsupervised biclustering (MoSBi), lipid network analysis (LINEX2), and pathway enrichment analysis confirmed alterations in amino acid-, lipid-, and nucleotide-related pathways. In tumor tissue, these pathways were significantly deregulated regarding gene and protein expression in two independent datasets, including actionable targets RRM2, GMPS, BCAT1, PYCR2, and NEU1. In vitro knockdown confirmed a functional role in proliferation and migration, as exemplified for PYCR2. Conclusions These findings demonstrate that serum metabolome profiling indicates deregulated metabolites and pathways during hepatocarcinogenesis. Our liquid biopsy approach accurately detects early-stage HCC outperforming currently recommended surveillance tools and facilitates identification of actionable candidates for chemoprevention. Impact and implications Deregulated cellular metabolism is a hallmark of cancer. In smaller studies, circulating metabolite profiles have been associated with HCC, although mainly in the context of fatty liver disease. Translation strategies for primary prevention or early detection are lacking. In this global study, we present an unsupervised landscape of the altered serum metabolome profile during hepatocarcinogenesis, independent of age, sex, and etiology. We provide a blood-based metabolite signature that accurately identifies early-stage HCC in a phase II biomarker study including independent validation. Further RRM2, GMPS, BCAT1, PYCR2, and NEU1 are identified in tumor tissue as actionable candidates for prevention. Our data provide the rationale for clinical trials testing liquid biopsy metabolome-based signatures for early HCC detection and the development of chemoprevention strategies.
Collapse
Affiliation(s)
- Kornelius Schulze
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- ERN-RARE-LIVER, Hamburg, Germany
| | - Tim Daniel Rose
- LipiTUM, Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Lorenz Adlung
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), and Center for Biomedical AI (bAIome), Hamburg, Germany
- Mildred Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manuela Peschka
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Newborn Screening and Metabolic Laboratory, Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Francesca Pagani
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Joao Gorgulho
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section of Pneumology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- University Cancer Center Hamburg–Hubertus Wald Tumorzentrum, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Thorben W. Fründt
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- ERN-RARE-LIVER, Hamburg, Germany
| | - Ismail Labgaa
- Division of Liver Diseases, Liver Cancer Program, Tisch Cancer Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Visceral Surgery, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Philipp K. Haber
- Division of Liver Diseases, Liver Cancer Program, Tisch Cancer Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carolin Zimpel
- Department of Medicine I, University Medical Center Schleswig-Holstein-Campus Lübeck, Germany
| | - Darko Castven
- Department of Medicine I, University Medical Center Schleswig-Holstein-Campus Lübeck, Germany
| | - Arndt Weinmann
- I. Department of Medicine, University Medical Center Mainz, Germany
| | - Teresa Garzia-Lezana
- Division of Liver Diseases, Liver Cancer Program, Tisch Cancer Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Moritz Waldmann
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Renné
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- Center for Thrombosis and Hemostasis (CTH), Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Hannah Voß
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manuela Moritz
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dorian Orlikowski
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hartmut Schlüter
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan Baumbach
- Chair of Computational Systems Biology, University of Hamburg, 22607 Hamburg, Germany
| | - Myron Schwartz
- Recanati Miller Transplant Institute, The Icahn School of Medicine at Mount Sinai Hospital, New York, NY, USA
| | - Ansgar W. Lohse
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- ERN-RARE-LIVER, Hamburg, Germany
| | - Samuel Huber
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- ERN-RARE-LIVER, Hamburg, Germany
| | - Bruno Sangro
- Liver Unit, Clinica Universidad de Navarra-IDISNA and CIBEREHD, Pamplona, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBEREHD), Carlos III National Institute of Health, Madrid, Spain
| | - Rocio I.R. Macias
- Center for the Study of Liver and Gastrointestinal Diseases (CIBEREHD), Carlos III National Institute of Health, Madrid, Spain
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, IBSAL, Salamanca, Spain
| | - Laura Izquierdo-Sanchez
- Department of Liver and Gastrointestinal Diseases, Biogipuzkoa Health Research Institute–Donostia University Hospital, University of the Basque Country (UPV/EHU), CIBEREHD, Donostia-San Sebastian, Spain
| | - Jesus M. Banales
- Department of Liver and Gastrointestinal Diseases, Biogipuzkoa Health Research Institute–Donostia University Hospital, University of the Basque Country (UPV/EHU), CIBEREHD, Donostia-San Sebastian, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Henning Wege
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- ERN-RARE-LIVER, Hamburg, Germany
| | - Jens U. Marquardt
- Department of Medicine I, University Medical Center Schleswig-Holstein-Campus Lübeck, Germany
- I. Department of Medicine, University Medical Center Mainz, Germany
| | - Augusto Villanueva
- Division of Liver Diseases, Liver Cancer Program, Tisch Cancer Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Hematology and Medical Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Josch Konstantin Pauling
- LipiTUM, Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Johann von Felden
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- ERN-RARE-LIVER, Hamburg, Germany
| |
Collapse
|
68
|
BharathwajChetty B, Kumar A, Deevi P, Abbas M, Alqahtani A, Liang L, Sethi G, Liu L, Kunnumakkara AB. Gut microbiota and their influence in brain cancer milieu. J Neuroinflammation 2025; 22:129. [PMID: 40312370 PMCID: PMC12046817 DOI: 10.1186/s12974-025-03434-2] [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: 11/21/2024] [Accepted: 04/01/2025] [Indexed: 05/03/2025] Open
Abstract
Microbial communities are not simply remnants of the past but dynamic entities that continuously evolve under the selective pressures of nature, reflecting the intricate and adaptive processes of evolution. The microbiota residing in the various regions of the human body has numerous roles in different physiological processes such as nutrition, metabolism, immune regulation, etc. In the zeal of achieving empirical insights into the ambit of the gut microbiome, the research over the years led to the revelation of reciprocal interaction between the gut microbiome and the cognitive functioning of the human body. Dysbiosis in the gut microbial composition disturbs the homeostatic cognitive functioning of the human body. This dysbiosis has been associated with various chronic diseases, including brain cancer, such as glioma, glioblastoma, etc. This review explores the mechanistic role of dysbiosis-mediated progression of brain cancers and their subtypes. Moreover, it demonstrates the regulatory role of microbial metabolites produced by the gut microbiota, such as short-chain fatty acids, amino acids, lipids, etc., in the tumour progression. Further, we also provide valuable insights into the microbiota mediating the efficiency of therapeutic regimens, thereby leveraging gut microbiota as potential biomarkers and targets for improved treatment outcomes.
Collapse
Affiliation(s)
- Bandari BharathwajChetty
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Aviral Kumar
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Pranav Deevi
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
- International Joint M. Tech Degree in Food Science and Technology, Department of Chemical Engineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha, 61421, Saudi Arabia
| | - Athba Alqahtani
- Research Centre, King Fahad Medical City, Riyadh, 11525, Saudi Arabia
| | - Liping Liang
- Guangzhou Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
- NUS Centre for Cancer Research, Yong Loo Lin Scool of Medicine, National University of Singapore, Singapore, 117699, Singapore.
| | - Le Liu
- Integrated Clinical Microecology Center, Shenzhen Hospital, Southern Medical University, Shenzhen, 518000, China.
- Department of Gastroenterology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India.
- International Joint M. Tech Degree in Food Science and Technology, Department of Chemical Engineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India.
| |
Collapse
|
69
|
Gao P, Zhou C, Yang H, Deng S, Jiang B, Wang Z, Di J, Su X. Comparison of circulating immune signatures across different consensus molecular subtypes (CMS) and between left- and right-sided lesions in stage II colorectal cancer using single-cell mass cytometry. J Transl Med 2025; 23:496. [PMID: 40312697 PMCID: PMC12044728 DOI: 10.1186/s12967-025-06481-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 04/11/2025] [Indexed: 05/03/2025] Open
Abstract
BACKGROUND Patients with stage II colorectal cancer (CRC) show considerable variability in prognosis. Circulating immune cells play a vital role in systemic tumor surveillance. This study aimed to determine the clinical significance of the frequency and phenotype of circulating immune cell subsets in patients with stage II CRC. METHODS We applied a 37-marker-cell-lineage-agnostic panel to perform single-cell mass cytometry on peripheral blood mononuclear cells (PBMCs) from 73 patients with stage II CRC and 21 patients with stage III CRC. To categorize the stage II patients into consensus molecular subtypes (CMS), we performed RNA sequencing on tumor and adjacent normal tissues from 51 of the 73 patients. We then compared the immune cell phenotypes and frequencies based on tumor location and CMS classification in patients with stage II CRC. Wilcoxon test was employed to compare the mean frequencies of cell clusters between different tumor locations. Krustal-Wallis analysis with post-hoc Dunn test was used to assess differences across multiple CMS groups. RESULTS Stage II CRC patients with left- and right-sided tumors, as well as in different CMS groups, exhibit significantly different tumor characteristics. Single-cell mass cytometry revealed profound interpatient variability in immune cell subpopulation distribution and phenotypes in PBMCs in stage II CRC. We identified unique T:monocyte complexes in the peripheral blood of patients with stage II CRC, with significantly higher frequencies in these patients compared to those with stage III CRC. Left- and right-sided stage II CRCs differed in peripheral immunity. Patients with right-sided CRC had significantly higher frequencies of circulating CD8+CD27-CD28- immunosenescent T cell subsets compared to patients with left-sided CRC. Significant variations were observed in the subpopulations of the T:monocyte complex, T cells, and monocytes across different CMS groups Patients with CMS1 tumors (immune-active) exhibited significantly higher frequencies of CD4+ central memory and CD8+ terminal effector T cell: classical monocyte complexes, as well as CD8+ terminal effector T cells in the circulation. CONCLUSIONS The frequency and phenotype of circulating immune cells are influenced by tumor side and CMS subtypes in stage II CRC. These observations provide a basis for further investigation into the mechanisms linking tumors and systemic immunity.
Collapse
Affiliation(s)
- Pin Gao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Chuanyong Zhou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Hong Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Shunyu Deng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Beihai Jiang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Zaozao Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Jiabo Di
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing, 100142, China.
| | - Xiangqian Su
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing, 100142, China.
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
70
|
Bhosale A, Meenakshi S, Narapaka PK, Richa C, Kalaiselvan V, Dhingra S, Kumar N, Rajesh R, Murti K. Assessment of patterns and predictors of medical device-related adverse events among oncology patients: a cross-sectional study at a tertiary care hospital. Expert Rev Med Devices 2025; 22:477-487. [PMID: 40159686 DOI: 10.1080/17434440.2025.2486471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 03/21/2025] [Accepted: 03/22/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND This study aims to identify patterns and predictors of medical device-related adverse events (MDAEs) among radiotherapy patients. Understanding MDAEs is crucial for optimizing patient safety during radiotherapy. METHODOLOGY An observational study conducted from August to December 2023 involved 139 patients undergoing radiotherapy. Demographics, clinical characteristics, and AE reports were collected from patient datasheets and analyzed using SPSS Version 28. RESULTS Study findings revealed that patients with head and neck cancer were significantly associated with higher rates of skin (OR: 3.56, CI: 1.59-7.96) and mucous membrane reactions. Specific dose ranges, particularly 800-2800 cGy, also predict mucous membrane reactions (OR: 11.12, CI: 3.42-36.1). Furthermore, smokeless habits significantly influenced both skin (OR: 6.04, CI: 1.99-18.3) and mucous membrane reactions (OR: 8.77, CI: 2.57-29.9). In contrast, head and neck cancer patients had reduced likelihoods of pharynx reactions (OR: 0.37, CI: 0.13-1.00), particularly with doses between 2801 and 4800 cGy (OR: 0.45, CI: 0.96-21.6). CONCLUSION This study identified a few significant predictors for the occurrence of various types of MDAEs among patients undergoing radiotherapy. Reporting MDAEs can prevent adverse effects caused by medical devices and enhance radiotherapy safety.
Collapse
Affiliation(s)
- Ashutosh Bhosale
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research, Hajipur, India
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal, India
| | - Sarasa Meenakshi
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research, Hajipur, India
| | - Pavan Kumar Narapaka
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research, Hajipur, India
| | - Chauhan Richa
- Department of Radiation Oncology, Mahavir Cancer Sansthan, Patna, India
| | - V Kalaiselvan
- Indian Pharmacopeia Commission, Ministry of Health and Family Welfare, Government of India, Ghaziabad, India
| | - Sameer Dhingra
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research, Hajipur, India
- Regional Training Centre, Materiovigilance Programme of India, National Institute of Pharmaceutical Education and Research, Hajipur, India
| | - Nitesh Kumar
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, India
| | - Radhakrishnan Rajesh
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal, India
| | - Krishna Murti
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research, Hajipur, India
- Regional Training Centre, Materiovigilance Programme of India, National Institute of Pharmaceutical Education and Research, Hajipur, India
| |
Collapse
|
71
|
Suleiman H, Emerson A, Wilson PM, Mulligan KA, Ladner RD, LaBonte MJ. Harnessing nucleotide metabolism and immunity in cancer: a tumour microenvironment perspective. FEBS J 2025; 292:2155-2172. [PMID: 39308084 PMCID: PMC12062787 DOI: 10.1111/febs.17278] [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: 04/30/2024] [Revised: 08/06/2024] [Accepted: 09/09/2024] [Indexed: 05/11/2025]
Abstract
The tumour microenvironment (TME) is a dynamic nexus where cancer cell metabolism and the immune system intricately converge, with nucleotide metabolism (NM) playing a pivotal role. This review explores the critical function of NM in cancer cell proliferation and its profound influence on the TME and immune landscape. NM is essential for DNA and RNA synthesis and is markedly upregulated in cancer cells to meet the demands of rapid growth. This metabolic rewiring fuels cancer progression, but also shapes the TME, impacting the function and viability of immune cells. The altered nucleotide milieu in the TME can suppress immune response, aiding cancer cell evasion from immune surveillance. Drug discoveries in the field of NM have revealed different therapeutic strategies, including inhibitors of nucleotide synthesis and drugs targeting salvage pathways, which are discussed thoroughly in this review. Furthermore, the emerging strategy of combining NM-targeted therapies with immunotherapies is emphasised, particularly their effect on sensitising tumours to immune checkpoint inhibitors and enhancing overall treatment efficacy. The Human Genome Project paved the way for personalised medicine, countering the established 'one size fits all' approach to cancer treatment. Advances in understanding the TME and NM have spurred interest in personalised therapeutic strategies. This review highlights the potential of leveraging individual tumour metabolic profiles to guide treatment selection, aiming to optimise efficacy and minimise adverse effects. The strategic importance of targeting NM in cancer therapy and its synergistic potential with immunotherapies offers a path towards more effective and personalised cancer treatments.
Collapse
Affiliation(s)
- Hadil Suleiman
- Patrick G Johnston Centre for Cancer ResearchQueen's University BelfastUK
| | - Alexandra Emerson
- Patrick G Johnston Centre for Cancer ResearchQueen's University BelfastUK
| | | | | | - Robert D. Ladner
- Patrick G Johnston Centre for Cancer ResearchQueen's University BelfastUK
- CV6 Therapeutics (NI) LtdBelfastUK
| | - Melissa J. LaBonte
- Patrick G Johnston Centre for Cancer ResearchQueen's University BelfastUK
| |
Collapse
|
72
|
Hu Y, Xiao Y. Mechanism of atorvastatin in treating hepatocellular carcinoma: a study based on network pharmacology, molecular docking, and bioinformatics analysis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:5693-5703. [PMID: 39607545 DOI: 10.1007/s00210-024-03598-3] [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: 09/19/2024] [Accepted: 11/01/2024] [Indexed: 11/29/2024]
Abstract
Hepatocellular carcinoma (HCC) is a tumor with high morbidity and mortality. Current research suggests that statins may aid in its prevention and treatment, while studies on the associated mechanisms remain limited. Therefore, we aim to reveal the mechanism of atorvastatin treatment for HCC by using network pharmacology and bioinformatics methods. The databases SwissTargetPrediction, PharmMapper, and DrugBank were utilized to obtain targets of atorvastatin, while GSE169289, GSE135631, and GSE207435 were used to identify differentially expressed genes (DEGs) for HCC. The overlap between the two groups was used to identify atorvastatin's target for treating HCC. Following protein-protein interaction (PPI) analysis, hub genes were identified using Cytoscape software and LASSO analysis. The hub genes were further validated using data from The Cancer Genome Atlas (TCGA) and The Human Protein Atlas (HPA) databases. To evaluate the clinical significance of the hub genes, Kaplan-Meier (KM) survival analysis and Cox analysis were conducted. Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Set Enrichment Analysis (GSEA) were performed to investigate potential mechanisms. Finally, molecular docking analysis was performed to validate the interaction between atorvastatin and the hub genes. A total of 1948 DEGs of HCC and 380 targets of atorvastatin were identified, respectively. After taking the intersection, 79 genes were identified as potential targets of atorvastatin for HCC treatment. After multiple screening methods, CYP2C9 was ultimately identified as the hub gene. Analysis of data from TCGA and HPA databases showed reduced expression of CYP2C9 in HCC tissues. KM and Cox analysis showed a favorable prognosis for HCC patients with high CYP2C9 expression. KEGG and GSEA indicated that metabolism of xenobiotics by cytochrome P450, and PPAR signaling pathway could be the potential mechanisms for atorvastatin in treating HCC. Molecular docking analysis revealed that atorvastatin binds to CYP2C9 with a binding energy of - 8.837, indicating highly stable binding. CYP2C9 is associated with the prognosis of HCC patients and could serve as a potential target for atorvastatin treatment in HCC.
Collapse
Affiliation(s)
- Youwen Hu
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Yangyang Xiao
- Department of Gerontology, Jiangxi University of Traditional Chinese Medicine Affiliated Hospital, No 445, Bayi Avenue, Nanchang, 330006, Jiangxi Province, China.
| |
Collapse
|
73
|
Shi X, Askari Rizvi SF, Yang Y, Liu G. Emerging nanomedicines for macrophage-mediated cancer therapy. Biomaterials 2025; 316:123028. [PMID: 39693782 DOI: 10.1016/j.biomaterials.2024.123028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/22/2024] [Accepted: 12/13/2024] [Indexed: 12/20/2024]
Abstract
Tumor-associated macrophages (TAMs) contribute to tumor progression by promoting angiogenesis, remodeling the tumor extracellular matrix, inducing tumor invasion and metastasis, as well as immune evasion. Due to the high plasticity of TAMs, they can polarize into different phenotypes with distinct functions, which are primarily categorized as the pro-inflammatory, anti-tumor M1 type, and the anti-inflammatory, pro-tumor M2 type. Notably, anti-tumor macrophages not only directly phagocytize tumor cells, but also present tumor-specific antigens and activate adaptive immunity. Therefore, targeted regulation of TAMs to unleash their potential anti-tumor capabilities is crucial for improving the efficacy of cancer immunotherapy. Nanomedicine serves as a promising vehicle and can inherently interact with TAMs, hence, emerging as a new paradigm in cancer immunotherapy. Due to their controllable structures and properties, nanomedicines offer a plethora of advantages over conventional drugs, thus enhancing the balance between efficacy and toxicity. In this review, we provide an overview of the hallmarks of TAMs and discuss nanomedicines for targeting TAMs with a focus on inhibiting recruitment, depleting and reprogramming TAMs, enhancing phagocytosis, engineering macrophages, as well as targeting TAMs for tumor imaging. We also discuss the challenges and clinical potentials of nanomedicines for targeting TAMs, aiming to advance the exploitation of nanomedicine for cancer immunotherapy.
Collapse
Affiliation(s)
- Xueying Shi
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular, Imaging and Translational Medicine, School of Public Health, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China
| | - Syed Faheem Askari Rizvi
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular, Imaging and Translational Medicine, School of Public Health, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China; Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, 54000, Punjab, Pakistan
| | - Yinxian Yang
- School of Pharmaceutical Sciences, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China.
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics Center for Molecular, Imaging and Translational Medicine, School of Public Health, Xiamen University, No. 4221 South Xiang'an Road, Xiang'an District, Xiamen, 361102, China.
| |
Collapse
|
74
|
Zhong J, Shi S, Peng W, Cui H, Sun X. HMGN2 accelerates the proliferation and cell cycle progression of glioblastoma by regulating CDC20 expression. Genes Dis 2025; 12:101433. [PMID: 40092489 PMCID: PMC11907455 DOI: 10.1016/j.gendis.2024.101433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 08/05/2024] [Accepted: 08/25/2024] [Indexed: 03/19/2025] Open
Abstract
Gliomas represent the most common primary malignant intracranial tumors in adults. Despite recent advances in treatment, the prognosis of patients with glioblastoma remains poor. Epigenetic abnormalities, the hallmarks of various types of cancer, contribute to the dysregulated expression of cancer-related genes. Post-translational modification of histones plays a pivotal role in cancer development and progression by modulating gene transcription, chromatin remodeling, and nuclear structure. Therefore, further exploration of the molecular mechanisms of epigenetic regulation in gliomas and the identification of superior therapeutic targets are required. High-mobility group nucleosomal-binding domain 2 (HMGN2) participates in the epigenetic regulation of genes through histone modification and exhibits significant differential expression between glioma and normal tissues. However, the effect of HMGN2 on gliomas and its underlying mechanisms remain unclear. This study aimed to elucidate these uncertainties by demonstrating that HMGN2 significantly promotes the proliferation of glioma cells. HMGN2 binds to histones and promotes the stability of H3K27ac acetylation in the cell division cycle 20 (CDC20) promoter region, enhancing the transcriptional activity of CDC20 and increasing the proliferation of glioma cells. Moreover, we found that CDC20 expression was negatively correlated with the survival time of patients with glioma. These results suggest that targeting epigenetic regulation, such as the HMGN2/CDC20 axis, may provide a novel direction for the treatment of gliomas.
Collapse
Affiliation(s)
- Jiacheng Zhong
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Shuang Shi
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Wen Peng
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400716, China
- Jinfeng Laboratory, Chongqing 401329, China
| | - Hongjuan Cui
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400716, China
- Jinfeng Laboratory, Chongqing 401329, China
| | - Xiaochuan Sun
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
75
|
Shen Q, Yang K, Li Q, Qin T, Yu Y, Hong H, Yao H, Xu X. Nanoparticles (NPs)-mediated silencing of GSTP1 expression to reverse chemoresistance for effective breast cancer therapy. J Colloid Interface Sci 2025; 685:38-48. [PMID: 39827759 DOI: 10.1016/j.jcis.2025.01.107] [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: 09/10/2024] [Revised: 01/11/2025] [Accepted: 01/12/2025] [Indexed: 01/22/2025]
Abstract
Chemotherapy remains the primary treatment modality for breast cancer (BCa) patients. However, chemoresistance commonly arises in clinical settings, contributing to poor prognosis. The development of chemoresistance is a dynamic and complex process involving the activation of oncogenes and inactivation of tumor suppressor genes. In this work, we utilized the RNA-sequencing (RNA-seq) technology to analyze the gene expression profiles of primary and recurrent tumor samples from BCa patients received the postoperative standard chemotherapy with doxorubicin (DOX), and identified glutathione S-transferase P1 (GSTP1) as a key factor in regulating chemoresistance. Molecular mechanistic studies revealed that high GSTP1 expression could not only impair the cytotoxicity of DOX by catalyzing the conjugation of reductive glutathione (GSH) with DOX, but also block the c-Jun NH2-terminal kinase (JNK) pathway to promote the proliferation via up-regulating anti-apoptotic B-cell lymphoma-2 (Bcl-2) expression. Given the severe side effects of DOX and the potential of RNA interference (RNAi) technology to silence target gene expression, we developed an endosomal pH-responsive nanoparticle (NP) platform for systemic co-delivery of DOX and GSTP1 siRNA (siGSTP1), and demonstrated its efficacy in reversing chemoresistance and suppressing the growth of DOX-resistant BCa tumors.
Collapse
Affiliation(s)
- Qian Shen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China
| | - Ke Yang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China
| | - Qingjian Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China
| | - Tao Qin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China
| | - Yunfang Yu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China
| | - Huangming Hong
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Department of Medical Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610041, PR China.
| | - Herui Yao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China.
| | - Xiaoding Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China; Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, PR China.
| |
Collapse
|
76
|
Xu Y, Fu G, Chen X, He R, Fang Q, Tan X, Xu K, Li H, Tan J, Fang M, Fu K, Huang Q, Xiao S. Integrated single-nuclear RNA sequencing analysis reveals distinct characteristics of mucinous adenocarcinoma in right-sided colon cancer. Int J Biol Macromol 2025; 309:142744. [PMID: 40180102 DOI: 10.1016/j.ijbiomac.2025.142744] [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: 11/18/2024] [Revised: 02/28/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025]
Abstract
Mucinous adenocarcinoma (MAC) is a unique histological subtype of colorectal cancer (CRC), which usually occurs in the right-sided colon with poor prognosis. Our previous studies reveled unique clinicopathological characteristic of MAC, but the distinct molecular features and tumor microenvironment (TME) characteristics remain clarify systematically. In this study, we conducted a single nuclear RNA sequencing (snRNA-seq) analysis for CRC tissues with different histological subtypes, including MAC, partial mucinous adenocarcinoma (pMAC) and non-specific adenocarcinoma (AC). Our results show that MAC has a unique transcriptome profile and a distinct single-cell characteristic. It exhibited a higher degree of tumor purity but a lower composition of TME. MAC had a distinct cell-cell communication microenvironment and a particular evolutionary trajectory. The EpithelialCells of MAC closely interacted with fibroblasts, endothelial cells, and mural cells clusters. Furthermore, molecular functional characteristics analysis revealed that MAC enriched EpithelialCells, Fibroblasts, and Endothelial cells clusters have a high active in glycolysis, inflammation, and angiogenesis respectively. This comprehensive study first demonstrated that MAC is a unique histological subtype of right-sided CRC in the single cell level, and elucidated its tumor microenvironment composition and biological function characteristics, which will help us better understand the intrinsic feature of MAC.
Collapse
Affiliation(s)
- Yunhua Xu
- The First Affiliated Hospital, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China; The First Affiliated Hospital, Institute of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Guang Fu
- The First Affiliated Hospital, Department of Gastrointestinal Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Xiguang Chen
- The First Affiliated Hospital, Department of Gastrointestinal Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Rongfang He
- The First Affiliated Hospital, Department of pathology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Qing Fang
- The First Affiliated Hospital, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China; The First Affiliated Hospital, Department of Gastrointestinal Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Xiangwen Tan
- The First Affiliated Hospital, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China; The Second Affiliated Hospital, Department of hepatological Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Kunming Xu
- The First Affiliated Hospital, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China; The First Affiliated Hospital, Department of Gastrointestinal Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Hongming Li
- The First Affiliated Hospital, Department of Gastrointestinal Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Jie Tan
- The First Affiliated Hospital, Department of Gastrointestinal Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Min Fang
- The First Affiliated Hospital, Department of Education and Training, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Kai Fu
- Institute of Molecular Precision Medicine, Hunan Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China.
| | - Qiulin Huang
- The First Affiliated Hospital, Department of Gastrointestinal Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China.
| | - Shuai Xiao
- The First Affiliated Hospital, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China; The First Affiliated Hospital, Institute of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China; The First Affiliated Hospital, Department of Gastrointestinal Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China.
| |
Collapse
|
77
|
Tian C, Yang S, Zhang C, Zhu R, Chen C, Wang X, Zhang D, Sun Q, Xu H, Nie H, Zhang Y, Ji D, Tang J, Jin K, Sun Y. Dual Role of CRABP2 in Colorectal Cancer: Oncogenesis via Nuclear RB1 and Cytoplasmic AFG3L2/SLC25A39 Axis, While Limiting Liver Metastasis through Cytoplasmic AFG3L2/PINK1/Parkin-Mediated Mitophagy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2500552. [PMID: 40305785 DOI: 10.1002/advs.202500552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/31/2025] [Indexed: 05/02/2025]
Abstract
Colorectal cancer (CRC) progression and metastasis involve numerous regulatory factors. Among these, cellular retinoic acid-binding protein 2 (CRABP2) has been implicated as both a tumor activator and suppressor. Here, it is aimed to clarify the role of CRABP2 in CRC growth and metastasis and explore the underlying molecular mechanisms mediating its cellular functions. Using both in vitro and in vivo models, including a colonocyte-specific CRABP2 conditional knockout mouse model (Crabp2ΔIEC) and a subcutaneous tumorigenesis assay in BALB/c nude mice, it is shown that nuclear CRABP2 enhances tumor growth by interacting with and downregulating the tumor suppressor RB1, whereas cytoplasmic CRABP2 suppresses CRC liver metastasis by interacting with AFG3L2 and promoting mitophagy. In addition, the AFG3L2-SLC25A39 axis is identified as a distinct mechanism by which cytoplasmic CRABP2 increases mitochondrial glutathione stability to promote cell proliferation independent of the nuclear RB1 pathway. Notably, analysis of tissue from CRC patients reveals that CRABP2 protein has distinct prognostic implications and functional roles in the progression and metastasis of CRC dependent on its subcellular localization. Ultimately, by elucidating the role of CRABP2 in CRC, it is aimed to provide new insight into disease pathogenesis and inform the development of therapeutic interventions.
Collapse
Affiliation(s)
- Chuanxin Tian
- Department of General Surgery, Colorectal Institute of Nanjing Medical University, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Sheng Yang
- Department of General Surgery, Colorectal Institute of Nanjing Medical University, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Chuan Zhang
- Department of General Surgery, Colorectal Institute of Nanjing Medical University, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Renzhong Zhu
- Institute of Translational Medicine, Medical College, Yangzhou University, No.136 Jiangyang Road, Yangzhou, 210029, China
| | - Chen Chen
- Department of General Surgery, Colorectal Institute of Nanjing Medical University, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Xiaowei Wang
- Department of General Surgery, Colorectal Institute of Nanjing Medical University, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Dongsheng Zhang
- Department of General Surgery, Colorectal Institute of Nanjing Medical University, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Qingyang Sun
- Department of General Surgery, Colorectal Institute of Nanjing Medical University, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Hengjie Xu
- Department of General Surgery, Colorectal Institute of Nanjing Medical University, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Hongxu Nie
- Department of General Surgery, Colorectal Institute of Nanjing Medical University, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Yue Zhang
- Department of General Surgery, Colorectal Institute of Nanjing Medical University, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Dongjian Ji
- Department of General Surgery, Colorectal Institute of Nanjing Medical University, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Junwei Tang
- Department of General Surgery, Colorectal Institute of Nanjing Medical University, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Kangpeng Jin
- Department of General Surgery, Colorectal Institute of Nanjing Medical University, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Yueming Sun
- Department of General Surgery, Colorectal Institute of Nanjing Medical University, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, 210029, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 210029, China
| |
Collapse
|
78
|
Yang Y, Zhang S, Wu Z, Li W, Sun X, Xuan Y, Hang T, Xu L, Chen X. Crystal structures of Kif2A complexed with WDR5 reveal the structural plasticity of WIN-S7 sites. Acta Biochim Biophys Sin (Shanghai) 2025. [PMID: 40302551 DOI: 10.3724/abbs.2025066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025] Open
Abstract
Chromosome congression and spindle assembly are essential for genomic stability and proper cell division, with deficiencies in these processes linked to tumorigenesis. WD repeat-containing protein 5 (WDR5), a core component of the mixed lineage leukemia (MLL) methyltransferase complex, directly binds to kinesin family member 2A (Kif2A) to regulate these mitotic events. Despite the importance of this interaction, its structural basis for Kif2A recognition by WDR5 remains unclear. Here, we determine the crystal structure of WDR5 in complex with a Kif2A-derived peptide (residues 114-122) at a resolution of 1.85 Å. Structural analysis reveals that Kif2A engages both the WIN and S7 sites of WDR5 via Arg117 and Ser121, with Ser121 forming hydrogen bonds with WDR5 Tyr191 and Lys259, driving Tyr191 rotation and opening the S7 pocket. Additional structures of WDR5 complexed with truncated or mutated Kif2A peptides and a WDR5 Y191F variant highlight the dynamic nature of Tyr191. Notably, anti-WDR5 compounds exhibit a similar binding mode at the WDR5 WIN-S7 site. The results of mutagenesis combined with isothermal titration calorimetry (ITC) assays underscore the critical roles of Arg117 and Ser121 in mediating the binding of Kif2A to WDR5. In summary, our findings provide atomic-level insights into the molecular mechanisms underlying the non-canonical mitotic function of the MLL/WDR5 complex and highlight WIN-S7 sites as promising therapeutic targets for diseases associated with chromosomal instability, such as cancers.
Collapse
Affiliation(s)
- Yang Yang
- School of Life Sciences, Anhui University, Hefei 230601, China
| | - Shuting Zhang
- School of Life Sciences, Anhui University, Hefei 230601, China
| | - Zhangyu Wu
- School of Life Sciences, Anhui University, Hefei 230601, China
| | - Wenwen Li
- School of Life Sciences, Anhui University, Hefei 230601, China
| | - Xuefang Sun
- School of Life Sciences, Anhui University, Hefei 230601, China
| | - Yumi Xuan
- Center for Human Tissues and Organs Degeneration, Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 581055, China
| | - Tianrong Hang
- School of Life Sciences, Anhui University, Hefei 230601, China
| | - Li Xu
- Institute of Bio-Architecture and Bio-Interactions (IBABI), Shenzhen Medical Academy of Research and Translation (SMART), Shenzhen 518107, China
| | - Xuemin Chen
- School of Life Sciences, Anhui University, Hefei 230601, China
| |
Collapse
|
79
|
Skaličková M, Abramenko N, Charnavets T, Vellieux F, Leischner Fialová J, Kučnirová K, Kejík Z, Masařík M, Martásek P, Pacak K, Pacák T, Jakubek M. Interaction of Selected Anthracycline and Tetracycline Chemotherapeutics with Poly(I:C) Molecules. ACS OMEGA 2025; 10:15935-15946. [PMID: 40321536 PMCID: PMC12044458 DOI: 10.1021/acsomega.4c05483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 02/11/2025] [Accepted: 04/09/2025] [Indexed: 05/08/2025]
Abstract
Despite the natural ability of the immune system to recognize cancer and, in some patients, even to eliminate it, cancer cells have acquired numerous evading mechanisms. With the increasing knowledge and focus shifting from targeting rapidly proliferating cells with chemotherapy to modulating the immune system, there have been recent efforts to integrate (e.g., simultaneously or sequentially) various therapeutic approaches. Combining the oncolytic activity of some chemotherapeutics with immunostimulatory molecules, so-called chemoimmunotherapy, is an attractive strategy. An example of such an immunostimulatory molecule is polyinosinic:polycytidylic acid [Poly(I:C)], a synthetic analogue of double-stranded RNA characterized by rapid nuclease degradation hampering its biological activity. This study investigated the possible interactions of tetracycline and anthracycline chemotherapeutics with different commercial Poly(I:C) molecules and protection against nuclease degradation. Fluorescence spectroscopy and circular dichroism revealed an interaction of all of the selected chemotherapeutics with Poly(I:C)s and the ability of doxycycline and minocycline to prolong the resistance to RNase cleavage, respectively. The partial protection was observed in vitro as well.
Collapse
Affiliation(s)
- Markéta Skaličková
- BIOCEV,
First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Department
of Paediatrics and Inherited Metabolic Disorders, First Faculty of
Medicine, Charles University and General
University Hospital, 120
00 Prague, Czech
Republic
| | - Nikita Abramenko
- BIOCEV,
First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Department
of Paediatrics and Inherited Metabolic Disorders, First Faculty of
Medicine, Charles University and General
University Hospital, 120
00 Prague, Czech
Republic
| | - Tatsiana Charnavets
- Institute
of Biotechnology of the Czech Academy of Sciences, BIOCEV, 252
50 Vestec, Czech
Republic
| | - Frédéric Vellieux
- BIOCEV,
First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Department
of Paediatrics and Inherited Metabolic Disorders, First Faculty of
Medicine, Charles University and General
University Hospital, 120
00 Prague, Czech
Republic
| | | | - Kateřina Kučnirová
- BIOCEV,
First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Department
of Paediatrics and Inherited Metabolic Disorders, First Faculty of
Medicine, Charles University and General
University Hospital, 120
00 Prague, Czech
Republic
| | - Zdeněk Kejík
- BIOCEV,
First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Department
of Paediatrics and Inherited Metabolic Disorders, First Faculty of
Medicine, Charles University and General
University Hospital, 120
00 Prague, Czech
Republic
| | - Michal Masařík
- BIOCEV,
First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Department
of Paediatrics and Inherited Metabolic Disorders, First Faculty of
Medicine, Charles University and General
University Hospital, 120
00 Prague, Czech
Republic
- Department
of Physiology, Faculty of Medicine, Masaryk
University, Kamenice 5, 625 00 Brno, Czech Republic
- Department
of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno CZ-625
00, Czech Republic
| | - Pavel Martásek
- Department
of Paediatrics and Inherited Metabolic Disorders, First Faculty of
Medicine, Charles University and General
University Hospital, 120
00 Prague, Czech
Republic
| | - Karel Pacak
- Section on
Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute
of Child Health and Human Development, National
Institutes of Health, Building 10, Room 1-3140, 10 Center Drive, Bethesda, Maryland 20892, United States
| | - Tomáš Pacák
- TumorSHOT, Italská 2581/67, Vinohrady,
Praha 2, Prague 120 00, Czech Republic
| | - Milan Jakubek
- BIOCEV,
First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Department
of Paediatrics and Inherited Metabolic Disorders, First Faculty of
Medicine, Charles University and General
University Hospital, 120
00 Prague, Czech
Republic
| |
Collapse
|
80
|
Delmas D, Mialhe A, Cotte AK, Connat JL, Bouyer F, Hermetet F, Aires V. Lipid metabolism in cancer: Exploring phospholipids as potential biomarkers. Biomed Pharmacother 2025; 187:118095. [PMID: 40311223 DOI: 10.1016/j.biopha.2025.118095] [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: 02/12/2025] [Revised: 04/03/2025] [Accepted: 04/24/2025] [Indexed: 05/03/2025] Open
Abstract
Aberrant lipid metabolism is increasingly recognized as a hallmark of cancer, contributing to tumor growth, metastatic dissemination, and resistance to therapy. Cancer cells reprogram key metabolic pathways-including de novo lipogenesis, lipid uptake, and phospholipid remodeling-to sustain malignant progression and adapt to microenvironmental demands. This review summarizes current insights into the role of lipid metabolic reprogramming in oncogenesis and highlights recent advances in lipidomics that have revealed cancer type- and stage-specific lipid signatures with diagnostic and prognostic relevance. We emphasize the dual potential of lipid metabolic pathways-particularly those involving phospholipids-as sources of clinically relevant biomarkers and therapeutic targets. Enzymes and transporters involved in these pathways have emerged as promising candidates for both diagnostic applications and pharmacological intervention. We also examine persistent challenges hindering the clinical translation of lipid-based approaches, including analytical variability, insufficient biological validation, and the lack of standardized integration into clinical workflows. Furthermore, the review explores strategies to overcome these barriers, highlighting the importance of incorporating lipidomics into multi-omics frameworks, supported by advanced computational tools and AI-driven analytics, to decipher the complexity of tumor-associated metabolic networks. We discuss how such integrative approaches can facilitate the identification of actionable metabolic targets, improve the specificity and robustness of lipid-based biomarkers, and enhance patient stratification in the context of precision oncology.
Collapse
Affiliation(s)
- Dominique Delmas
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France; Centre de Lutte Contre le Cancer Georges François Leclerc Center, Dijon F-21000, France; Inserm UMS58 - Biologie Santé Dijon (BioSanD), Dijon F-21000, France.
| | - Aurélie Mialhe
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France
| | - Alexia K Cotte
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France
| | - Jean-Louis Connat
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France
| | - Florence Bouyer
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France
| | - François Hermetet
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France
| | - Virginie Aires
- Université Bourgogne Europe, Dijon F-21000, France; Inserm Research Center UMR1231 - Therapies and Immune Response in Cancers Team, Bioactive Molecules and Health Research Group, Dijon F-21000, France
| |
Collapse
|
81
|
Zhou Z, Ma Y, Zhang D, Ji R, Wang Y, Zhao J, Ma C, Zhu H, Shen H, Jiang X, Niu Y, Lu J, Zhang B, Tu L, Zhang H, Ma X, Chen P. Microbiome and fragmentation pattern of blood cell-free DNA and fecal metagenome enhance colorectal cancer micro-dysbiosis and diagnosis analysis: a proof-of-concept study. mSystems 2025:e0027625. [PMID: 40298367 DOI: 10.1128/msystems.00276-25] [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: 02/24/2025] [Accepted: 04/02/2025] [Indexed: 04/30/2025] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer, and it can be prevented by performing early screening. As a hallmark of cancer, the human microbiome plays important roles in the occurrence and development of CRC. Recently, the blood microbiome has been proposed as an effective diagnostic tool for various diseases, yet its performance on CRC deserves further exploration. In this study, 133 human feces and 120 blood samples are collected, including healthy individuals, adenoma patients, and CRC patients. The blood cfDNA and fecal genome are subjected to shotgun metagenome sequencing. After removing human sequences, the microbial sequences in blood are analyzed. Based on the differential microbes and functions, random forest (RF) models are constructed for adenoma and CRC diagnosis. The results show that alterations of blood microbial signatures can be captured under low coverage (even at 3×). RF diagnostic models based on blood microbial markers achieve high area under the curve (AUC) values for adenoma patients (0.8849) and CRC patients (0.9824). When the fragmentation pattern is combined with microbial and KEGG markers, higher AUC values are obtained. Furthermore, compared to the blood microbiome, the fecal microbiome shows a different community composition, whereas their changes in KEGG pathways are similar. Pathogenic bacteria Fusobacterium nucleatum (F. nucleatum) in feces increased gradually from the healthy group to the adenoma and CRC groups. Additionally, F. nucleatum in feces and blood shows a positive correlation in CRC patients. Cumulatively, the integration of blood microbiome and fragmentation pattern is promising for CRC diagnosis.IMPORTANCEThe cell-free DNA of the human microbiome can enter the blood and can be used for cancer diagnosis, whereas its diagnostic potential in colorectal cancer and association with gut microbiome has not been explored. The microbial sequences in blood account for less than 1% of the total sequences. The blood microbial composition, KEGG functions, and fragmentation pattern are different among healthy individuals, adenoma patients, and CRC patients. Machine learning models based on these differential characteristics achieve high diagnostic accuracy, especially when they are integrated with fragmentation patterns. The great difference between fecal and blood microbiomes indicates that microbial sequences in blood may originate from various organs. Therefore, this study provides new insights into the community composition and functions of the blood microbiome of CRC and proposes an effective non-invasive diagnostic tool.
Collapse
Affiliation(s)
- Zhongkun Zhou
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, China
| | - Yunhao Ma
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, China
| | - Dekui Zhang
- The Second Hospital of Lanzhou University, Lanzhou, China
| | - Rui Ji
- The First Hospital of Lanzhou University, Lanzhou, China
| | - Yiqing Wang
- The First Hospital of Lanzhou University, Lanzhou, China
| | - Jianfang Zhao
- The Third People's Hospital of Gansu Province, Lanzhou, China
| | - Chi Ma
- The Second Hospital of Lanzhou University, Lanzhou, China
| | - Hongmei Zhu
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, China
| | - Haofei Shen
- The First Hospital of Lanzhou University, Lanzhou, China
| | - Xinrong Jiang
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, China
| | - Yuqing Niu
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, China
| | - Juan Lu
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, China
| | - Baizhuo Zhang
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, China
| | - Lixue Tu
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, China
| | - Hua Zhang
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, China
| | - Xin Ma
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, China
| | - Peng Chen
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
82
|
Thangavelu PU, Lin CY, Forouz F, Tanaka K, Dray E, Duijf PHG. The RB protein: more than a sentry of cell cycle entry. Trends Mol Med 2025:S1471-4914(25)00088-7. [PMID: 40300971 DOI: 10.1016/j.molmed.2025.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/28/2025] [Accepted: 04/02/2025] [Indexed: 05/01/2025]
Abstract
Genomic instability is a hallmark of cancer. It fuels cancer progression and therapy resistance. As 'the guardian of the genome', the tumor suppressor protein p53 protects against genomic damage. Canonically, the retinoblastoma protein (RB) is 'the sentry of cell cycle entry', as it dictates whether a cell enters the cell cycle to divide. However, the RB pathway also controls myriad non-canonical cellular processes, including metabolism, stemness, angiogenesis, apoptosis, and immune surveillance. We discuss how frequent RB pathway inactivation and underlying mechanisms in cancers affect these processes. We focus on RB's - rather than p53's - 'guardian of the genome' functions in DNA replication, DNA repair, centrosome duplication, chromosome segregation, and chromatin organization. Finally, we review therapeutic strategies, challenges, and opportunities for targeting RB pathway alterations in cancer.
Collapse
Affiliation(s)
- Pulari U Thangavelu
- Frazer Institute, University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - Cheng-Yu Lin
- Frazer Institute, University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - Farzaneh Forouz
- School of Pharmacy, Faculty of Health and Behavioural Sciences, University of Queensland, Brisbane, QLD, Australia
| | - Kozo Tanaka
- Department of Molecular Oncology, Institute of Development, Aging and Cancer (IDAC), Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Eloïse Dray
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA; Mays Cancer Center at UT Health San Antonio MD Anderson, San Antonio, TX, USA; Greehey Children's Cancer Research Institute, San Antonio, TX, USA
| | - Pascal H G Duijf
- Centre for Cancer Biology, Clinical and Health Sciences, University of South Australia & SA Pathology, Adelaide, SA, Australia; School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
83
|
Adlakha YK, Chhabra R. The human microbiome: redefining cancer pathogenesis and therapy. Cancer Cell Int 2025; 25:165. [PMID: 40296128 PMCID: PMC12039184 DOI: 10.1186/s12935-025-03787-x] [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: 09/27/2024] [Accepted: 04/11/2025] [Indexed: 04/30/2025] Open
Abstract
The human microbiome has always been an important determinant of health and recently, its role has also been described in cancer. The altered microbiome could aid cancer progression, modulate chemoresistance and significantly alter drug efficacy. The broad implications of microbes in cancer have prompted researchers to investigate the microbe-cancer axis and identify whether modifying the microbiome could sensitize cancer cells for therapy and improve the survival outcome of cancer patients. The preclinical data has shown that enhancing the number of specific microbial species could restore the patients' response to cancer drugs and the microbial biomarkers may play a vital role in cancer diagnostics. The elucidation of detailed interactions of the human microbiota with cancer would not only help identify the novel drug targets but would also enhance the efficacy of existing drugs. The field exploring the emerging roles of microbiome in cancer is at a nascent stage and an in-depth scientific perspective on this topic would make it more accessible to a wider audience. In this review, we discuss the scientific evidence connecting the human microbiome to the origin and progression of cancer. We also discuss the potential mechanisms by which microbiota affects initiation of cancer, metastasis and chemoresistance. We highlight the significance of the microbiome in therapeutic outcome and evaluate the potential of microbe-based cancer therapy.
Collapse
Affiliation(s)
- Yogita K Adlakha
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Sector-125, Noida, Uttar Pradesh, 201303, India.
| | - Ravindresh Chhabra
- Department of Biochemistry, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India.
| |
Collapse
|
84
|
Skrabalak I, Rajtak A, Malachowska B, Skrzypczak N, Skalina KA, Guha C, Kotarski J, Okla K. Therapy resistance: Modulating evolutionarily conserved heat shock protein machinery in cancer. Cancer Lett 2025; 616:217571. [PMID: 39986370 DOI: 10.1016/j.canlet.2025.217571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/16/2025] [Accepted: 02/17/2025] [Indexed: 02/24/2025]
Abstract
Therapy resistance is a major barrier to achieving a cure in cancer patients, often resulting in relapses and mortality. Heat shock proteins (HSPs) are a group of evolutionarily conserved proteins that play a prominent role in the progression of cancer and drug resistance. HSP synthesis is upregulated in cancer cells, facilitating adaptation to various tumor microenvironment (TME) stressors, including nutrient deprivation, exposure to DNA-damaging agents, hypoxia, and immune responses. In this review, we present background information about HSP-mediated cancer therapy resistance. Within this context, we emphasize recent progress in the understanding of HSP machinery, exploring the therapeutic potential of HSPs in cancer treatment.
Collapse
Affiliation(s)
- Ilona Skrabalak
- The First Department of Oncologic Gynecology and Gynecology, Medical University of Lublin, Lublin, Poland
| | - Alicja Rajtak
- The First Department of Oncologic Gynecology and Gynecology, Medical University of Lublin, Lublin, Poland; IOA, 3 Lotnicza St, 20-322 Lublin, Poland
| | - Beata Malachowska
- Department of Radiation Oncology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
| | - Natalia Skrzypczak
- Department of Pathology and Clinical Laboratories, University of Michigan, Ann Arbor, MI, USA
| | - Karin A Skalina
- Department of Radiation Oncology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
| | - Chandan Guha
- Department of Radiation Oncology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
| | - Jan Kotarski
- The First Department of Oncologic Gynecology and Gynecology, Medical University of Lublin, Lublin, Poland
| | - Karolina Okla
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA; IOA, 3 Lotnicza St, 20-322 Lublin, Poland.
| |
Collapse
|
85
|
Shao Q, Gao HY, Wang ZY, Qian YL, Chen WX. Construction of a novel five programmed cell death-related gene signature as a promising prognostic model for triple negative breast cancer. PeerJ 2025; 13:e19359. [PMID: 40313394 PMCID: PMC12045267 DOI: 10.7717/peerj.19359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 04/02/2025] [Indexed: 05/03/2025] Open
Abstract
Background Triple negative breast cancer (TNBC) is a more aggressive subtype of breast cancer that usually progresses rapidly, develops drug resistance, metastasis, and relapses, and remains a challenge for clinicians to treat. Programmed cell death (PCD), a conserved mechanism of cell suicide controlled by various pathways, contributed to carcinogenesis and cancer progression. Nevertheless, the prognostic significance of PCD-related genes in TNBC remains largely unclear, and more accurate prognostic models are urgently needed. Methods Gene expression profiles and clinical information of TNBC patients were obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) database. Least absolute shrinkage and selection operator (LASSO) and multivariate Cox regression analysis were used to establish the PCD-related gene signature. Kaplan-Meier plotter, receiver operating characteristic curves, and nomogram were applied to validate the prognostic value of the gene signature. Gene set enrichment analysis was carried out to investigate the pathways and molecular functions. Results Five PCD-related genes including SEPTIN3, SCARB1, CHML, SYNM, and COL5A3 were identified to establish the PCD-related risk score for TNBC patients. Patients stratified into high-risk or low-risk group showed significantly different survival outcome, immune infiltration, and drug susceptibility. Kaplan-Meier and receiver operating characteristic curves showed a good performance for survival prediction in different cohorts. Gene set enrichment analysis revealed that the five-gene signature was associated with tumor metabolism, cancer cell proliferation, invasion and metastasis, and tumor microenvironment. Nomogram including the five-gene signature was established. Conclusion A novel five PCD-related gene signature and nomogram could be used for prognostic prediction in TNBC. The present work might offer useful insights in digging sensitive and effective biomarkers for TNBC prognosis prediction and establishing accurate prognostic model in clinical management.
Collapse
Affiliation(s)
- Quanfeng Shao
- Department of Breast Surgery, Changzhou No. 2 People’s Hospital, The Third Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Hai-yan Gao
- Department of Breast Surgery, Changzhou Cancer Hospital, Changzhou, Jiangsu, China
| | - Zi-ying Wang
- Department of Breast Surgery, Changzhou No. 2 People’s Hospital, The Third Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Yu-ling Qian
- Kangda Clinical College, Kangda College of Nanjing Medical University, Lianyungang, Jiangsu, China
| | - Wei-xian Chen
- Department of Breast Surgery, Changzhou No. 2 People’s Hospital, The Third Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
- Post-doctoral Working Station, Changzhou No. 2 People’s Hospital, The Third Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| |
Collapse
|
86
|
Mohamed NM, Mohamed RH, Kennedy JF, Elhefnawi MM, Hamdy NM. A comprehensive review and in silico analysis of the role of survivin (BIRC5) in hepatocellular carcinoma hallmarks: A step toward precision. Int J Biol Macromol 2025; 311:143616. [PMID: 40306500 DOI: 10.1016/j.ijbiomac.2025.143616] [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: 02/13/2025] [Revised: 04/25/2025] [Accepted: 04/27/2025] [Indexed: 05/02/2025]
Abstract
Hepatocellular carcinoma (HCC) is a complex malignancy driven by the dysregulation of multiple cellular pathways. Survivin, a key member of the inhibitor of apoptosis (IAP) family, plays a central role in HCC tumorigenesis and progression. Despite significant research, a comprehensive understanding of the contributions of survivin to the hallmarks of cancer, its molecular network, and its potential as a therapeutic target remains incomplete. In this review, we integrated bioinformatics analysis with an extensive literature review to provide deeper insights into the role of survivin in HCC. Using bioinformatics tools such as the Human Protein Atlas, GEPIA, STRING, TIMER, and Metascape, we analyzed survivin expression and its functional associations and identified the top 20 coexpressed genes in HCC. These include TK1, SPC25, SGO2, PTTG1, PRR11, PLK1, NCAPH, KPNA2, KIF2C, KIF11, HJURP, GTSE1, FOXM1, CEP55, CENPA, CDCA3, CDC45, CCNB2, CCNB1 and CTD-2510F5.4. Our findings also revealed significant protein-protein interactions among these genes, which were enriched in pathways associated with the FOXM1 oncogenic signaling cascade, and biological processes such as cell cycle regulation, mitotic checkpoints, and diseases such as liver neoplasms. We also discussed the involvement of survivin in key oncogenic pathways, including the PI3K/AKT, WNT/β-catenin, Hippo, and JAK/STAT3 pathways, and its role in modulating cell cycle checkpoints, apoptosis, and autophagy. Furthermore, we explored its interactions with the tumor microenvironment, particularly its impact on immune modulation through myeloid-derived suppressor cells (MDSCs), tumor-associated macrophages, and natural killer cell function in HCC. Additionally, we highlighted its involvement in alkylglycerone phosphate synthase (AGPS)-mediated lipid reprogramming and identified important gaps in the survivin network that warrant further investigation. This review also examined the role of survivin in cancer stemness, inflammation, and virally mediated hepatocarcinogenesis. We evaluated its potential as a diagnostic, prognostic, predictive, and pharmacodynamic biomarker in HCC, emphasizing its relevance in precision medicine. Finally, we summarized emerging survivin-targeted therapeutics and ongoing clinical trials, underscoring the need for novel strategies to effectively target survivin in HCC.
Collapse
Affiliation(s)
- Nermin M Mohamed
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Abassia, 11566 Cairo, Egypt
| | - Rania Hassan Mohamed
- Department of Biochemistry, Faculty of Science, Ain Shams University, Abassia, 11566 Cairo, Egypt
| | - John F Kennedy
- Chembiotech Laboratories, Kyrewood House, Tenbury Wells, Worcestershire, United Kingdom
| | - Mahmoud M Elhefnawi
- Biomedical Informatics and Chemoinformatics Group, Informatics and Systems Department, National Research Centre, Cairo, Egypt.
| | - Nadia M Hamdy
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Abassia, 11566 Cairo, Egypt.
| |
Collapse
|
87
|
Rodríguez-Martínez A, Giraldo-Ruiz L, Ramos MC, Luque I, Ribeiro D, Postigo-Corrales F, Alburquerque-González B, Montoro-García S, Arroyo-Rodríguez AB, Conesa-Zamora P, Hurtado AM, Luengo-Gil G, Pérez-Sánchez H. Discovery of Z1362873773: a novel fascin inhibitor from a large chemical library for colorectal cancer. Sci Rep 2025; 15:14906. [PMID: 40295602 PMCID: PMC12037858 DOI: 10.1038/s41598-025-96457-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 03/28/2025] [Indexed: 04/30/2025] Open
Abstract
Metastasis is one of the leading causes of cancer-related death worldwide. Fascin, a protein that bundles actin filaments to produce protrusions in cancer cells, plays a significant role in the enhancement of cell migration. This protein has been shown that the overexpression of this protein is related to the appearance of different types of cancer, such as colorectal cancer. In this study, we conducted in silico screening of the Enamine library, a compound library with a broad chemical space. Using a ligand-based virtual screening approach based on the pharmacophore model of G2, we identified the predicted inhibitors. First, these compounds were validated by physicochemical analysis. Differential scanning calorimetry (DSF) was used to study the binding between the predicted compounds and fascin protein, followed by an F-actin bundling assay to determine which compounds inhibited the bundling function of fascin. Z1362873773, which exhibited binding to fascin and inhibited F-actin bundling, was further tested in cell cultures to assess its effects on cancer cell viability and migration as well as in organoid models to evaluate potential cytotoxicity. Finally, we established a protocol that can be applied to discover anti-fascin agents from diverse compound libraries. A new molecule has been identified with considerable fascin inhibitory and migration-arresting capacity, which may lead to the development of new therapies to treat cancer.
Collapse
Affiliation(s)
- Alejandro Rodríguez-Martínez
- Structural Bioinformatics and High Performance Computing Research Group (BIO-HPC), UCAM Universidad Católica de Murcia (UCAM), HiTech Innovation Hub, Murcia, 30107, Spain
- Health Sciences PhD Program, Universidad Católica de Murcia UCAM, Campus de los Jerónimos nº135, Guadalupe, Murcia, 30107, Spain
| | - Lucía Giraldo-Ruiz
- Departamento de Química Física, Instituto de Biotecnología y Unidad de Excelencia de Química aplicada a Biomedicina y Medioambiente, Universidad de Granada, Campus Fuentenueva s/n, Granada, 18071, Spain
| | - María C Ramos
- Fundación MEDINA, PTS Health Sciences Technology Park, Avenida del Conocimiento 34, Granada, 18016, Spain
| | - Irene Luque
- Departamento de Química Física, Instituto de Biotecnología y Unidad de Excelencia de Química aplicada a Biomedicina y Medioambiente, Universidad de Granada, Campus Fuentenueva s/n, Granada, 18071, Spain
| | - Diogo Ribeiro
- Genetics, Molecular Pathology and Rare Diseases Research Group, Universidad Católica de Murcia (UCAM), Guadalupe, 30107, Spain
| | - Fátima Postigo-Corrales
- Genetics, Molecular Pathology and Rare Diseases Research Group, Universidad Católica de Murcia (UCAM), Guadalupe, 30107, Spain
| | - Begoña Alburquerque-González
- Genetics, Molecular Pathology and Rare Diseases Research Group, Universidad Católica de Murcia (UCAM), Guadalupe, 30107, Spain
| | - Silvia Montoro-García
- Genetics, Molecular Pathology and Rare Diseases Research Group, Universidad Católica de Murcia (UCAM), Guadalupe, 30107, Spain
| | - Ana Belén Arroyo-Rodríguez
- Molecular Pathology and Pharmacogenetics Research Group, Instituto Murciano de Investigación Biosanitaria (IMIB), Hospital General Universitario Santa Lucía, Cartagena, 30202, Spain
| | - Pablo Conesa-Zamora
- Genetics, Molecular Pathology and Rare Diseases Research Group, Universidad Católica de Murcia (UCAM), Guadalupe, 30107, Spain
- Molecular Pathology and Pharmacogenetics Research Group, Instituto Murciano de Investigación Biosanitaria (IMIB), Hospital General Universitario Santa Lucía, Cartagena, 30202, Spain
| | - Ana María Hurtado
- Genetics, Molecular Pathology and Rare Diseases Research Group, Universidad Católica de Murcia (UCAM), Guadalupe, 30107, Spain
| | - Ginés Luengo-Gil
- Genetics, Molecular Pathology and Rare Diseases Research Group, Universidad Católica de Murcia (UCAM), Guadalupe, 30107, Spain.
- Molecular Pathology and Pharmacogenetics Research Group, Instituto Murciano de Investigación Biosanitaria (IMIB), Hospital General Universitario Santa Lucía, Cartagena, 30202, Spain.
| | - Horacio Pérez-Sánchez
- Structural Bioinformatics and High Performance Computing Research Group (BIO-HPC), UCAM Universidad Católica de Murcia (UCAM), HiTech Innovation Hub, Murcia, 30107, Spain.
| |
Collapse
|
88
|
Huang D, Cai H, Huang H. Serine metabolism in tumor progression and immunotherapy. Discov Oncol 2025; 16:628. [PMID: 40295433 PMCID: PMC12037972 DOI: 10.1007/s12672-025-02358-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 04/09/2025] [Indexed: 04/30/2025] Open
Abstract
Serine plays a vital role in various metabolic processes including the synthesis of proteins and other amino acids, which are essential for the cell proliferation and growth. Cancer cells either absorb exogenous serine or produce it through the serine synthesis pathway, enabling the generation of intracellular glycine and one-carbon units, which are crucial for nucleotide biosynthesis. This metabolic process, referred to as serine-glycine-one-carbon (SGOC) metabolism, is essential for tumorigenesis and exhibits considerable complexity and clinical significance. Enzymes involved in the SGOC pathway are linked to tumor growth, metastasis, and resistance to therapies. The SGOC pathway is a vital metabolic network that facilitates cell survival and proliferation, especially in aggressive cancers. Understanding how this network is regulated is crucial for tackling tumor heterogeneity and recurrence. This review emphasizes recent advancements in understanding the roles and effects of the SGOC metabolic pathway in the context of cancer progression. Additionally, it outlines the complex influences of the SGOC metabolic pathway on the tumor microenvironment (TME), offering potential strategies to enhance cancer immunotherapy.
Collapse
Affiliation(s)
- Dong Huang
- Oncology Hematology Department, Fengdu General Hospital, Fengdu County, Chongqing, 408200, China
| | - Hui Cai
- Oncology Hematology Department, Fengdu General Hospital, Fengdu County, Chongqing, 408200, China
| | - HaiYu Huang
- Oncology Hematology Department, Fengdu General Hospital, Fengdu County, Chongqing, 408200, China.
| |
Collapse
|
89
|
Amereh M, Seyfoori A, Shojaei S, Lane S, Zhao T, Shokrollahi Barough M, Lum JJ, Walter P, Akbari M. Tumoroid Model Reveals Synergistic Impairment of Metabolism by Iron Chelators and Temozolomide in Chemo-Resistant Patient-derived Glioblastoma Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2412505. [PMID: 40285641 DOI: 10.1002/advs.202412505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 03/18/2025] [Indexed: 04/29/2025]
Abstract
Chemoresistance poses a significant clinical challenge in managing glioblastoma (GBM), limiting the long-term success of traditional treatments. Here, a 3D tumoroid model is used to investigate the metabolic sensitivity of temozolomide (TMZ)-resistant GBM cells to iron chelation by deferoxamine (DFO) and deferiprone (DFP). This work shows that TMZ-resistant GBM cells acquire stem-like characteristics, higher intracellular iron levels, higher expression of aconitase, and elevated reliance on oxidative phosphorylation and proteins associated with iron metabolism. Using a microphysiological model of GBM-on-a-chip consisting of extracellular matrix (ECM)-incorporated tumoroids, this work demonstrates that the combination of iron chelators with TMZ induces a synergistic effect on an in vitro tumoroid model of newly diagnosed and recurrent chemo-resistant patient-derived GBM and reduced their size and invasion. Investigating downstream metabolic variations reveal reduced intracellular iron, increased reactive oxygen species (ROS), upregulated hypoxia-inducible factor-1α, reduced viability, increased autophagy, upregulated ribonucleotide reductase (RRM2), arrested proliferation, and induced cell death in normoxic TMZ-resistant cells. Hypoxic cells, while showing similar results, display reduced responses to iron deficiency, less blebbing, and an induced autophagic flux, suggesting an adaptive mechanism associated with hypoxia. These findings show that co-treatment with iron chelators and TMZ induces a synergistic effect, making this combination a promising GBM therapy.
Collapse
Affiliation(s)
- Meitham Amereh
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC, V8P 5C2, Canada
| | - Amir Seyfoori
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC, V8P 5C2, Canada
| | - Shahla Shojaei
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| | - Sarah Lane
- Department of Biology, University of Victoria, BC, Canada
| | - Tian Zhao
- Trev and Joyce Deeley Research Centre, BC Cancer, Victoria, BC, V8R 6V5, Canada
| | - Mahdieh Shokrollahi Barough
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC, V8P 5C2, Canada
| | - Julian J Lum
- Trev and Joyce Deeley Research Centre, BC Cancer, Victoria, BC, V8R 6V5, Canada
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, V8W 2Y2, Canada
| | - Patrick Walter
- Department of Biology, University of Victoria, BC, Canada
| | - Mohsen Akbari
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC, V8P 5C2, Canada
- Terasaki Institute for Biomedical Innovations, Los Angeles, CA, 91367, USA
| |
Collapse
|
90
|
Tan X, He W, Chen T, Yang W, Huang D, Zhang H, Luo Y, Lu M, Zhang Z, Ji J, Liu H. Immune-related gene expression with colorectal cancer risk: a Mendelian randomization analysis. Discov Oncol 2025; 16:617. [PMID: 40285908 PMCID: PMC12033153 DOI: 10.1007/s12672-025-02291-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 04/01/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND The potential role of immune cells and the precise contributions of genes from various immune cells in the development of colorectal cancer (CRC) remains unclear. We aimed to investigate the potential causal relationships between specific immune cell genes and the development of CRC by performing Mendelian randomization (MR) analysis. METHODS The cis-expression quantitative trait loci (eQTLs) data for type-specific immune cells were used to perform MR analysis. Outcomes data were obtained from the latest Genome Wide Association Studies (GWAS), comprising 78,473 European ancestry CRC cases and 107,143 controls. A few sensitivity analyses, encompassing pleiotropy and colocalization analyses, along with the application of False Discovery Rate (FDR) correction, were carried out to mitigate potential biases. RESULTS A total of 395 genes were found to be associated with CRC. After FDR correction, a total of 47 genes across 14 immune cell types were identified. Notably, the FHL3 gene showed the strongest association with CRC in multiple immune cell types, including CD4 + naïve and central memory T cells (CD4NC), CD4 + T cells with an effector memory or central memory phenotype (CD4ET), CD8 + naïve and central memory T cells (CD8NC), CD8 + T cells with an effector memory phenotype (CD8ET), and NK recruiting cells (NKR). Sensitivity analyses and pleiotropy assessments excluded the bias due to weak instruments and linkage disequilibrium. Additionally, the associations were supported by strong evidence of colocalization with CRC. The expression of FHL3 in normal tissue was higher than that in the tumor tissue at the mRNA expression and single-cell levels, and low mRNA expression of FHL3 was associated with shorter survival time in CRC patients. CONCLUSIONS Our study provides a novel perspective on the potential causal link between the low expression of FHL3 gene and the risk of CRC. More evidence is essential to further reveal the biological mechanisms underlying this association.
Collapse
Affiliation(s)
- Xin Tan
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wenjun He
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Tao Chen
- Nanfang Hospital, Southern Medical University, Guangzhou, China
- Clinical Research Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weihao Yang
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Donghua Huang
- Department of Health Management, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hengyi Zhang
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yangyang Luo
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mengting Lu
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhenzhan Zhang
- Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Jianguang Ji
- Department of Public Health and Medicinal Administration, Faculty of Health Sciences, University of Macau, Macao, Macao SAR, China.
| | - Hao Liu
- Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
91
|
Wang J, Yuan T, Yang B, He Q, Zhu H. SDH defective cancers: molecular mechanisms and treatment strategies. Cell Biol Toxicol 2025; 41:74. [PMID: 40285898 PMCID: PMC12033202 DOI: 10.1007/s10565-025-10022-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 04/13/2025] [Indexed: 04/29/2025]
Abstract
Succinate dehydrogenase (SDH), considered as the linkage between tricarboxylic acid cycle (TCA cycle) and electron transport chain, plays a vital role in adenosine triphosphate (ATP) production and cell physiology. SDH deficiency is a notable characteristic in many cancers. Recent studies have pinpointed the dysregulation of SDH can directly result its decreased catalytic activity and the accumulation of oncometabolite succinate, promoting tumor progression in different perspectives. This article expounds the various types of SDH deficiency in tumors and the corresponding pathological features. In addition, we discuss the mechanisms through which defective SDH fosters carcinogenesis, pioneering a categorization of these mechanisms as being either succinate-dependent or independent. Since SDH-deficient and cumulative succinate are regarded as the typical features of some cancers, like gastrointestinal stromal tumors, pheochromocytomas and paragangliomas, we summarize the presented medical management of SDH-deficient tumor patients in clinical and preclinical, identifying the potential strategies for future cancer therapeutics.
Collapse
Affiliation(s)
- Jiaer Wang
- Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Hangzhou, China
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310000, China
| | - Tao Yuan
- Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Hangzhou, China
| | - Bo Yang
- Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Hangzhou, China
- School of Medicine, Hangzhou City University, Hangzhou, 310015, China
| | - Qiaojun He
- Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Hangzhou, China.
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310020, China.
| | - Hong Zhu
- Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Hangzhou, China.
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310000, China.
| |
Collapse
|
92
|
Fiser O, Muller P. Role of HSF1 in cell division, tumorigenesis and therapy: a literature review. Cell Div 2025; 20:11. [PMID: 40287736 PMCID: PMC12034185 DOI: 10.1186/s13008-025-00153-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 04/08/2025] [Indexed: 04/29/2025] Open
Abstract
Heat shock factor 1 (HSF1) is the master orchestrator of the heat shock response (HSR), a critical process for maintaining cellular health and protein homeostasis. These effects are achieved through rapid expression of molecular chaperones, the heat shock proteins (HSPs), which ensure correct protein folding, repair, degradation and stabilization of multiprotein complexes. In addition to its role in the HSR, HSF1 influences the cell cycle, including processes such as S phase progression and regulation of the p53 pathway, highlighting its importance in cellular protein synthesis and division. While HSF1 activity offers neuroprotective benefits in neurodegenerative diseases, its proteome-stabilizing function may also reinforce tumorigenic transformation. HSF1 overexpression in many types of cancer reportedly enhances cell growth enables survival, alters metabolism, weakens immune response and promotes angiogenesis or epithelial-mesenchymal transition (EMT) as these cells enter a form of "HSF1 addiction". Furthermore, the client proteins of HSF1-regulated chaperones, particularly Hsp90, include numerous key players in classical tumorigenic pathways. HSF1 thus presents a promising therapeutic target for cancer treatment, potentially in combination with HSP inhibitors to alleviate typical initiation of HSR upon their use.
Collapse
Affiliation(s)
- Otakar Fiser
- Research Centre for Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute, Brno, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Petr Muller
- Research Centre for Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute, Brno, Czech Republic.
| |
Collapse
|
93
|
Huang X, Ji M, Shang X, Zhang H, Zhang X, Zhou J, Yin T. Smart on-demand drug release strategies for cancer combination therapy. J Control Release 2025; 383:113782. [PMID: 40294796 DOI: 10.1016/j.jconrel.2025.113782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 04/06/2025] [Accepted: 04/24/2025] [Indexed: 04/30/2025]
Abstract
In cancer therapy, enhancing therapeutic indices and patient compliance has been a central focus in recent drug delivery technology development. However, achieving a delicate balance between improving anti-tumor efficacy and minimizing toxicity to normal tissues remains a significant challenge. With the advent of smart on-demand drug release strategies, new opportunities have emerged. These strategies represent a promising approach to drug delivery, enabling precise control over the release of therapeutic agents in a programmed and spatiotemporal manner. Recent studies have focused on designing delivery systems capable of releasing multiple therapeutic agents sequentially, while achieving spatial resolution in vivo. Smart on-demand drug release strategies have demonstrated considerable potential in tumor combination therapy for achieving precision drug delivery and controlled release by responding to specific physiological signals or external physical stimuli in the tumor microenvironment. These strategies not only improve tumor targeting and reduce toxicity to healthy tissues but also enable sequential release in combination therapy, allowing multiple drugs to be released in a specific spatiotemporal order to enhance synergistic treatment effects. In this paper, we systematically reviewed the current research progress of smart on-demand drug release drug delivery strategies in anti-tumor combination therapy. We highlighted representative integrated drug delivery systems and discussed the challenges associated with their clinical application. Additionally, potential future research directions are proposed to further advance this promising field.
Collapse
Affiliation(s)
- Xiaolin Huang
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Mengfei Ji
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Xinyu Shang
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Hengchuan Zhang
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Xin Zhang
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Jianping Zhou
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China.
| | - Tingjie Yin
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China.
| |
Collapse
|
94
|
Zheng P, Hu Z, Shen Y, Gu L, Ouyang Y, Duan Y, Ji G, Dong B, Lin Y, Wen T, Tian Q, Hou Y, Zhou Q, Sun X, Chen X, Wang KL, Luo S, Wu S, Sun Y, Li M, Xiao L, Wu Q, Meng Y, Liu G, Wang Z, Bai X, Duan S, Ding Y, Bi Y, Wang Y, Li G, Liu X, Lu Z, Wu X, Tang Z, Xu D. PSAT1 impairs ferroptosis and reduces immunotherapy efficacy via GPX4 hydroxylation. Nat Chem Biol 2025:10.1038/s41589-025-01887-3. [PMID: 40281343 DOI: 10.1038/s41589-025-01887-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 03/14/2025] [Indexed: 04/29/2025]
Abstract
Tumor cells adapt to the inflammatory tumor microenvironment (TME) and develop resistance to immunotherapy, with ferroptosis being a major form of tumor cell death. However, the mechanisms by which tumor cells coordinate TME stimuli and their unique metabolic traits to evade ferroptosis and develop resistance to immunotherapy remain unclear. Here we showed that interferon-γ (IFNγ)-activated calcium/calmodulin-dependent protein kinase II phosphorylates phosphoserine aminotransferase 1 (PSAT1) at serine 337 (S337), allowing it to interact with glutathione peroxidase 4 (GPX4) and stabilize the protein, counteracting ferroptosis. PSAT1 elevates GPX4 stability by promoting α-ketoglutarate-dependent PHD3-mediated GPX4 proline 159 (P159) hydroxylation, disrupting its binding to HSC70 and inhibiting autophagy-mediated degradation. In mice, reconstitution of PSAT1 S337A or GPX4 P159A promotes ferroptosis and suppresses triple-negative breast cancer (TNBC) progression. Blocking PSAT1 pS337 with CPP elevates IFNγ-induced ferroptosis and enhances the efficacy of programmed cell death protein 1 (PD-1) antibodies in TNBC. Additionally, PSAT1-mediated GPX4 hydroxylation correlates with poor immunotherapy outcomes in patients with TNBC, highlighting PSAT1's noncanonical role in suppressing ferroptosis and immunotherapy sensitivity.
Collapse
Affiliation(s)
- Peixiang Zheng
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Zhiqiang Hu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Yuli Shen
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Lina Gu
- Department of Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yuan Ouyang
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Yuran Duan
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Guimei Ji
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Bofei Dong
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Yanni Lin
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Ting Wen
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Qi Tian
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Yueru Hou
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Qimin Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xue Sun
- Department of Surgical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xiaohan Chen
- Department of Surgical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | | | - Shudi Luo
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Shiqi Wu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Yuening Sun
- Department of Pharmacy, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Min Li
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Liwei Xiao
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Qingang Wu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Ying Meng
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Guijun Liu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Zheng Wang
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Xueli Bai
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shengzhong Duan
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Yuan Ding
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanli Bi
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, China
| | - Yuhao Wang
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Gaopeng Li
- Department of Colorectal Surgery and Oncology of the Second Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoguang Liu
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education) of the Second Affiliated Hospital and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhimin Lu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China
| | - Xiaohong Wu
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, China.
| | - Zhiyuan Tang
- Department of Pharmacy, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China.
| | - Daqian Xu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China.
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, China.
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, China.
| |
Collapse
|
95
|
Huang Z, Li L, Zhao X, Jin H, Shen M, Li B, Zeng Y, Zhang Q, Wang Q, Wang M, Yang L. scRNA-seq reveals that VEGF signaling mediates the response to neoadjuvant anlotinib combined with PD-1 blockade therapy in non-small cell lung cancer. J Transl Med 2025; 23:478. [PMID: 40281576 PMCID: PMC12032801 DOI: 10.1186/s12967-025-06485-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Accepted: 04/11/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Clinical trials have shown that neoadjuvant anlotinib combined with PD-1 blockade therapy can prolong the survival of patients with driver gene negative non-small cell lung cancer (NSCLC), but some patients fail to benefit from the combination therapy. METHODS To explore the potential drug resistance mechanism and predict the efficacy of neoadjuvant therapy in NSCLC patients, we used scRNA-seq to observe and analyze the dynamic changes of immune cells, stromal cells and cancer cells in NSCLC patients who received neoadjuvant combination therapy. We analyzed transcriptome data of ~ 47,000 single cells from 9 NSCLC patients, including 3 treatment naïve patients, 3 post-treatment patients with major pathological response (MPR), and 3 Non-MPR patients. Subsequently, the infiltration of immune cells was detected by immunohistochemistry and multiplex immunofluorescence in NSCLC. RESULTS In MPR patients, we found that neoadjuvant therapy reduced the expression of the T cell exhausted signature, reduced the transition of T_THEMIS cells to Tregs, and enhanced the positive feedback between CD4+ T cells and PAX5+ memory B cells. In Non-MPR patients, tumor-associated macrophages (TAMs) dampen therapeutic efficiency by being the hub of cell communication. TAMs and fibroblasts stimulate endothelial cells via VEGF, endothelial ZEB1 may up-regulate FLT1 (VEGFR) expression in response to anlotinib, and VEGFR+ endothelial cell signature can predict survival of NSCLC cohort in TCGA. In addition, PLA2G4A, the key enzyme in the VEGF pathway, was highly expressed in the tumor cells of Non-MPR patients after anlotinib treatment. In 135 NSCLC patients, we confirmed by immunohistochemistry that PLA2G4A was positively correlated with poor prognosis and Tregs infiltration. CONCLUSION In conclusion, VEGF signaling dependent dynamic changes in endothelial and epithelial cells are deeply involved in the formation of anlotinib resistance and immunosuppression phenotypes in NSCLC patients.
Collapse
Affiliation(s)
- Ziqi Huang
- Department of Immunology, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, China
| | - Li Li
- Department of Immunology, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, China
| | - Xiaohe Zhao
- Department of Immunology, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, China
| | - Haixia Jin
- Department of Immunology, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, China
| | - Meng Shen
- Department of Immunology, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, China
| | - Baihui Li
- Department of Esophageal Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Yu Zeng
- Department of Immunology, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, China
| | - Qinfen Zhang
- Department of Immunology, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, China
| | - Qiyu Wang
- Department of Immunology, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, China
| | - Meng Wang
- Department of Lung Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, China.
| | - Lili Yang
- Department of Immunology, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, China.
| |
Collapse
|
96
|
Borlongan MC, Rodriguez T, Putthanbut N, Wang H, Lee JY. Modeling of cancer stem cells and the tumor microenvironment Via NT2/D1 cells to probe pathology and treatment for cancer and beyond. Discov Oncol 2025; 16:605. [PMID: 40272656 PMCID: PMC12022208 DOI: 10.1007/s12672-025-02158-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 03/17/2025] [Indexed: 04/27/2025] Open
Abstract
INTRODUCTION Unique from the other tumor cells, tumorigenic cancer stem cells (CSCs) manifest as a subpopulation of cells within the tumor that exhibit genetic and phenotypic features and signaling processes, which escape traditional anti-oncogenic treatments, thereby triggering metastases and relapses of cancers. Critical to cancer biology is the crosstalk between CSCs and tumor microenvironment (TME), implicating a CSC-based cancer immunotherapy. Cognizant of CSCs' significant role in cancer pathology and treatment, finding a biological model that recapitulates CSCs and TME may allow a better understanding of tumor onset and progression for testing CSC-based therapies. In this review paper, we examined the CSC and TME characteristics of the human embryonal carcinoma NTERA-2 clonal cell line called NTERA-2 cl.D1 or NT2/D1 cells and discussed their potential utility for research and development of treatments for cancer and central nervous system (CNS) disorders. METHODS To probe our hypotheses that NT2/D1 cells display CSC and TME properties key to tumor development, which can serve as a screening platform to test cancer and CNS therapeutics, we conducted a literature review over a 10-year period (2014-2024), focusing on PUBMED and Science Direct published articles on cellular models of cancer, with emphasis on milestone research discoveries on NT2/D1 cells relevant to CSCs and TME. We categorized the studies under pre-clinical and clinical investigations in supporting the existence of CSC and TME features in NT2/D1 cells and providing a laboratory-to-clinic translational basis for cancer and CNS therapeutics. CONCLUSIONS NT2/D1 cells stand as a feasible biological model that recapitulates the crosstalk of CSCs and TME, which may critically contribute to our understanding of cancer and CNS biology and therapeutics. Designing therapeutics against CSCs' distinct self-renewal and differentiation capacities within the TME opens new avenues for treating cancers and CNS disorders.
Collapse
Affiliation(s)
- Mia C Borlongan
- California Northstate University College of Medicine, Elk Grove, CA, 95757, USA
| | - Thomas Rodriguez
- Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA
| | - Napasiri Putthanbut
- Center of Aging and Brain Repair, Department of Neurosurgery, University of South Florida, Tampa, FL, 33612, USA
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Hongbin Wang
- California Northstate University College of Pharmacy, Elk Grove, CA, 95757, USA
| | - Jea-Young Lee
- Center of Aging and Brain Repair, Department of Neurosurgery, University of South Florida, Tampa, FL, 33612, USA.
| |
Collapse
|
97
|
Wang LJ, Lei CL, Wang TA, Lin ZF, Feng SJ, Wei T, Li YQ, Shen MR, Li Y, Liao LF. Prognostic value of the preoperative systemic immune-inflammation nutritional index in patients with gastric cancer. World J Clin Oncol 2025; 16:102294. [PMID: 40290682 PMCID: PMC12019271 DOI: 10.5306/wjco.v16.i4.102294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/04/2024] [Accepted: 01/21/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) is the fifth most common cancer and the third leading cause of cancer-related deaths in China. Many patients with GC frequently experience symptoms related to the disease, including anorexia, nausea, vomiting, and other discomforts, and often suffer from malnutrition, which in turn negatively affects perioperative safety, prognosis, and the effectiveness of adjuvant therapeutic measures. Consequently, some nutritional indicators such as nutritional risk index (NRI), prognostic nutritional index (PNI), and systemic immune-inflammatory-nutritional index (SIINI) can be used as predictors of the prognosis of GC patients. AIM To examine the prognostic significance of PNI, NRI, and SIINI in postoperative patients with GC. METHODS A retrospective analysis was conducted on the clinical data of patients with GC who underwent surgical treatment at the Guangxi Medical University Cancer Hospital between January 2010 and December 2018. The area under the receiver operating characteristic (ROC) curve was assessed using ROC curve analysis, and the optimal cutoff values for NRI, PNI, and SIINI were identified using the You-Review-HTMLden index. Survival analysis was performed using the Kaplan-Meier method. In addition, univariate and multivariate analyses were conducted using the Cox proportional hazards regression model. RESULTS This study included a total of 803 patients. ROC curves were used to evaluate the prognostic ability of NRI, PNI, and SIINI. The results revealed that SIINI had superior predictive accuracy. Survival analysis indicated that patients with GC in the low SIINI group had a significantly better survival rate than those in the high SIINI group (P < 0.05). Univariate analysis identified NRI [hazard ratio (HR) = 0.68, 95% confidence interval (CI): 0.52-0.89, P = 0.05], PNI (HR = 0.60, 95%CI: 0.46-0.79, P < 0.001), and SIINI (HR = 2.10, 95%CI: 1.64-2.69, P < 0.001) as prognostic risk factors for patients with GC. However, multifactorial analysis indicated that SIINI was an independent risk factor for the prognosis of patients with GC (HR = 1.65, 95%CI: 1.26-2.16, P < 0.001). CONCLUSION Analysis of clinical retrospective data revealed that SIINI is a valuable indicator for predicting the prognosis of patients with GC. Compared with NRI and PNI, SIINI may offer greater application for prognostic assessment.
Collapse
Affiliation(s)
- Li-Jing Wang
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Cai-Lu Lei
- School of Pharmaceutical Science, Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Ting-An Wang
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Zhi-Feng Lin
- School of Pharmaceutical Science, Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Shi-Jie Feng
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Tao Wei
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Yan-Qin Li
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Meng-Ru Shen
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Yan Li
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Liu-Feng Liao
- Department of Pharmacy, Guangxi Medical University Cancer Hospital, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
98
|
Behrooz AB, Latifi-Navid H, Zolfaghari N, Piroozmand S, Pour-Rashidi A, Bourbour M, Jusheghani F, Aghaei M, Azarpira N, Mollasalehi F, Alamdar S, Nasimian A, Lotfi J, Shojaei S, Nazar E, Ghavami S. Metabolic reprogramming in glioblastoma: a rare case of recurrence to scalp metastasis. BJC REPORTS 2025; 3:27. [PMID: 40274950 PMCID: PMC12022025 DOI: 10.1038/s44276-025-00134-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 02/09/2025] [Accepted: 03/18/2025] [Indexed: 04/26/2025]
Abstract
BACKGROUND Glioblastoma (GB), an aggressive brain malignancy with a poor prognosis of 1.5-2 years, rarely exhibits extracranial metastasis (ECM). However, metabolic reprogramming has emerged as a key driver of GB progression and invasiveness. This study presents a rare case of recurrent GB with scalp metastasis, exploring how metabolic shifts enable GB cells to evade treatment and adapt to hostile environments, offering insights for developing innovative therapies. METHODS Tandem mass spectrometry (MS/MS) was employed to analyze amino acid profiles in both the recurrent and metastatic stages of GB. Systems biology approaches were used to uncover genetic alterations and metabolic reprogramming associated with the progression from recurrence to metastasis. RESULTS Our analysis revealed distinct amino acid utilization patterns in a patient with a molecular phenotype of wild-type IDH-1&2, TERT mutation, non-mutated BRAF and EGFR, and non-methylated MGMT. During recurrence and metastasis, significant differences in amino acid profiles were observed between blood and cerebrospinal fluid (CSF) samples. Additionally, protein-protein interaction (PPI) analysis identified key genomic drivers potentially responsible for the transition from recurrent to metastatic GB. CONCLUSIONS Beyond established risk factors such as craniotomy, biopsies, ventricular shunting, and radiation therapy, our findings suggest that metabolic reprogramming plays a crucial role in the transition from recurrent to metastatic GB. Targeting these metabolic shifts could provide new avenues for managing and preventing extracranial metastasis in GB, making this an important focus for future research.
Collapse
Affiliation(s)
- Amir Barzegar Behrooz
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB, Canada
| | - Hamid Latifi-Navid
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
- School of Biological Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
| | - Narges Zolfaghari
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Somayeh Piroozmand
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | | | - Mahsa Bourbour
- Department of Biotechnology, Alzahra University, Tehran, Iran
| | - Fatemeh Jusheghani
- Department of Biotechnology, Asu vanda Gene Industrial Research Company, Tehran, Iran
| | - Mahmoud Aghaei
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Negar Azarpira
- Shiraz Institute for Stem Cell and Regenerative Medicine, Shiraz University of Medical Science, Shiraz, Iran
- Transplant Research Center, Shiraz University of Medical Science, Shiraz, Iran
| | | | - Sedigheh Alamdar
- Clinical and Anatomical Pathology Department, Milad Hospital, Tehran, Iran
| | - Ahmad Nasimian
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB, Canada
| | - Jabar Lotfi
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- Growth and development research center, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahla Shojaei
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB, Canada
| | - Elham Nazar
- Department of Pathology, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB, Canada.
- Research Institute of Oncology and Hematology, Cancer Care Manitoba, University of Manitoba, Winnipeg, MB, Canada.
- Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
99
|
Klußmann M, Matijass M, Neundorf I. Impact of Mutational Status on Intracellular Effects of Cell-Permeable CaaX Peptides in Pancreatic Cancer Cells. Chembiochem 2025:e2401076. [PMID: 40270247 DOI: 10.1002/cbic.202401076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 03/31/2025] [Indexed: 04/25/2025]
Abstract
Prenyltransferases add a lipid group to the cysteine of a CaaX motif of proteins. This posttranslational modification enables proteins to attach to membranes where they are essential hubs for signaling, trafficking, and apoptosis. Recently, cell-permeable CaaX-peptides are developed as possible tools to interfere with the prenylation machinery. These peptides cause cytotoxic effects, particularly in KRas mutant pancreatic cancer cells (PANC-1) in which they also alter downstream signaling of Ras proteins. Herein, the aim is to get more clues about the relevance of the mutational status of KRas. Therefore, the activity of CaaX-peptides in KRas wildtype BxPC-3 and KRas mutated PANC-1 cells is compared. CaaX-peptides differently influence these two cell lines, although they internalize pretty much to the same extent. Indeed, an altered KRas plasma membrane localization in PANC-1 cells is observed, probably induced by disturbed KRas prenylation based on the presence of CaaX-peptides. The impact of CaaX-peptides on KRas signaling is likely dependent on the KRas mutation in PANC-1 cells in which they further trigger effects on KRas-dependent regulators, e.g., Neurofibromin -1 (NF1) and son of sevenless homolog 1 (SOS1). All in all, CaaX peptides are identified as promising tools for studying and manipulating the function of therapeutically important prenylated proteins.
Collapse
Affiliation(s)
- Merlin Klußmann
- Department of Chemistry and Biochemistry, Institute of Biochemistry, University of Cologne, Zuelpicher Str. 47a, 50674, Cologne, Germany
| | - Martin Matijass
- Department of Chemistry and Biochemistry, Institute of Biochemistry, University of Cologne, Zuelpicher Str. 47a, 50674, Cologne, Germany
| | - Ines Neundorf
- Department of Chemistry and Biochemistry, Institute of Biochemistry, University of Cologne, Zuelpicher Str. 47a, 50674, Cologne, Germany
| |
Collapse
|
100
|
Bai Y, Liu Q, Yang Y, Abudumijiti G, Huang Y, Chi J, Ji Y, Zhao Y, Liu B. Chemical reprogramming of liver cancer cells through targeting Aurora kinases. SCIENCE CHINA. LIFE SCIENCES 2025:10.1007/s11427-023-2795-2. [PMID: 40304920 DOI: 10.1007/s11427-023-2795-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 10/17/2024] [Indexed: 05/02/2025]
Affiliation(s)
- Yunfei Bai
- State Key Laboratory of Natural and Biomimetic Drugs, Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing Advanced Center of Cellular Homeostasis and Aging-Related Diseases, Institute of Advanced Clinical Medicine, College of Future Technology, Peking University, Beijing, 100871, China
- Plastech Pharmaceutical Technology Co., Ltd., Nanjing, 210031, China
| | - Qian Liu
- Department of Pathology, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215002, China
| | - Yang Yang
- State Key Laboratory of Natural and Biomimetic Drugs, Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing Advanced Center of Cellular Homeostasis and Aging-Related Diseases, Institute of Advanced Clinical Medicine, College of Future Technology, Peking University, Beijing, 100871, China
| | - Gulinigeer Abudumijiti
- State Key Laboratory of Natural and Biomimetic Drugs, Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing Advanced Center of Cellular Homeostasis and Aging-Related Diseases, Institute of Advanced Clinical Medicine, College of Future Technology, Peking University, Beijing, 100871, China
| | - Yanni Huang
- Plastech Pharmaceutical Technology Co., Ltd., Nanjing, 210031, China
| | - Jun Chi
- Plastech Pharmaceutical Technology Co., Ltd., Nanjing, 210031, China
| | - Yin Ji
- State Key Laboratory of Natural and Biomimetic Drugs, Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing Advanced Center of Cellular Homeostasis and Aging-Related Diseases, Institute of Advanced Clinical Medicine, College of Future Technology, Peking University, Beijing, 100871, China
- Nanjing Reju Therapeutics, Co. Ltd., Nanjing, 210000, China
| | - Yang Zhao
- State Key Laboratory of Natural and Biomimetic Drugs, Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing Advanced Center of Cellular Homeostasis and Aging-Related Diseases, Institute of Advanced Clinical Medicine, College of Future Technology, Peking University, Beijing, 100871, China.
- Plastech Pharmaceutical Technology Co., Ltd., Nanjing, 210031, China.
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China.
| | - Biao Liu
- Department of Pathology, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215002, China.
| |
Collapse
|