201
|
Song S, Fan Y, Zou G, Huo L, Kumar J, Li Y, Wang R, Dai E, Jin J, Scott AW, Shao S, Pizzi MP, Vykoukal JV, Katayama H, Hanash S, Calin GA, Zhang X, Lee MG, Wang Z, Lo YH, Gan Q, Waters RE, Yin F, Wang L, Cheng X, Ajani JA, Dhar SS. KAP1 promotes gastric adenocarcinoma progression by activating Hippo/YAP1 signaling via binding to HNRNPAB. Cancer Lett 2025; 621:217695. [PMID: 40189014 DOI: 10.1016/j.canlet.2025.217695] [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: 12/19/2024] [Revised: 03/31/2025] [Accepted: 04/03/2025] [Indexed: 04/15/2025]
Abstract
Gastric adenocarcinoma (GAC) remains a significant global health challenge, with over a million new cases annually. Peritoneal carcinomatosis (PC), detected in ∼20 % of cases at diagnosis and ∼45 % later, is uniformly fatal, with limited treatment options. This study investigated the role of KAP1 in GAC progression, focusing on its interaction with YAP1 and cancer stemness traits. Analysis of over 596 primary GACs and 72 PC samples revealed that high nuclear KAP1 expression correlates with poor prognosis. KAP1 knockdown reduced oncogenic activity and stemness traits in GAC cells. Mechanistically, KAP1 positively regulates YAP1 transcription by binding to its promoter and reducing H3K27ac levels. Mass spectrometry identified an interaction between KAP1 and HNRNPAB, further modulating YAP1 signaling. Expression of the KRAB domain of ZFP568 without its DNA-binding zinc fingers inhibited both KAP1 and YAP1 expression, significantly reducing colony formation and tumor growth in vivo. Additionally, emerging antisense oligonucleotides (ASOs) targeting KAP1 or YAP1 effectively suppressed mouse tumor progression. These findings establish KAP1 as a critical driver of tumor progression in GAC through YAP1 regulation and HNRNPAB interaction, highlighting its potential therapeutic target. This study advances our understanding and offers a preclinical framework to improve outcomes for GAC.
Collapse
Affiliation(s)
- Shumei Song
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yibo Fan
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gengyi Zou
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Longfei Huo
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Janani Kumar
- Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yuan Li
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Surgical Oncology and General Surgery, First Hospital of China Medical University, Shenyang, 110001, PR China
| | - Ruiping Wang
- Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Enyu Dai
- Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jiankang Jin
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ailing W Scott
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shan Shao
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Melissa Pool Pizzi
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jody V Vykoukal
- Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hiroyuki Katayama
- Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Samir Hanash
- Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - George A Calin
- Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xing Zhang
- Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Min Gyu Lee
- Molecular & Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zhenning Wang
- Department of Surgical Oncology and General Surgery, First Hospital of China Medical University, Shenyang, 110001, PR China
| | - Yuan-Hung Lo
- Molecular & Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Qiong Gan
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rebecca E Waters
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Feng Yin
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Linghua Wang
- Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiaodong Cheng
- Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Shilpa S Dhar
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
202
|
Furrer R, Handschin C. Biomarkers of aging: from molecules and surrogates to physiology and function. Physiol Rev 2025; 105:1609-1694. [PMID: 40111763 DOI: 10.1152/physrev.00045.2024] [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: 10/30/2024] [Revised: 01/10/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025] Open
Abstract
Many countries face an unprecedented challenge in aging demographics. This has led to an exponential growth in research on aging, which, coupled to a massive financial influx of funding in the private and public sectors, has resulted in seminal insights into the underpinnings of this biological process. However, critical validation in humans has been hampered by the limited translatability of results obtained in model organisms, additionally confined by the need for extremely time-consuming clinical studies in the ostensible absence of robust biomarkers that would allow monitoring in shorter time frames. In the future, molecular parameters might hold great promise in this regard. In contrast, biomarkers centered on function, resilience, and frailty are available at the present time, with proven predictive value for morbidity and mortality. In this review, the current knowledge of molecular and physiological aspects of human aging, potential antiaging strategies, and the basis, evidence, and potential application of physiological biomarkers in human aging are discussed.
Collapse
|
203
|
Hamza M, Wang S, Liu Y, Li K, Zhu M, Chen L. Unraveling the potential of bioengineered microbiome-based strategies to enhance cancer immunotherapy. Microbiol Res 2025; 296:128156. [PMID: 40158322 DOI: 10.1016/j.micres.2025.128156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 03/21/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025]
Abstract
The human microbiome plays a pivotal role in the field of cancer immunotherapy. The microbial communities that inhabit the gastrointestinal tract, as well as the bacterial populations within tumors, have been identified as key modulators of therapeutic outcomes, affecting immune responses and reprogramming the tumor microenvironment. Advances in synthetic biology have made it possible to reprogram and engineer these microorganisms to improve antitumor activity, enhance T-cell function, and enable targeted delivery of therapies to neoplasms. This review discusses the role of the microbiome in modulating both innate and adaptive immune mechanisms-ranging from the initiation of cytokine production and antigen presentation to the regulation of immune checkpoints-and discusses how these mechanisms improve the efficacy of immune checkpoint inhibitors. We highlight significant advances with bioengineered strains like Escherichia coli Nissle 1917, Lactococcus lactis, Bifidobacterium, and Bacteroides, which have shown promising antitumor efficacy in preclinical models. These engineered microorganisms not only efficiently colonize tumor tissues but also help overcome resistance to standard therapies by reprogramming the local immune environment. Nevertheless, several challenges remain, such as the requirement for genetic stability, effective tumor colonization, and the control of potential safety issues. In the future, the ongoing development of genetic engineering tools and the optimization of bacterial delivery systems are crucial for the translation of microbiome-based therapies into the clinic. This review highlights the potential of bioengineered microbiota as an innovative, personalized approach in cancer immunotherapy, bringing hope for more effective and personalized treatment options for patients with advanced malignancies.
Collapse
Affiliation(s)
- Muhammad Hamza
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuai Wang
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, China
| | - Yike Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Kun Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Motao Zhu
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, China; CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Lin Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
204
|
Sato Y. Immune Aging and Its Implication for Age-Related Disease Progression. Physiology (Bethesda) 2025; 40:0. [PMID: 39887318 DOI: 10.1152/physiol.00051.2024] [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: 11/11/2024] [Revised: 12/18/2024] [Accepted: 01/25/2025] [Indexed: 02/01/2025] Open
Abstract
As life expectancy increases globally, the prevalence and severity of age-related diseases have risen, significantly impacting patients' quality of life and increasing dependency on the healthcare system. Age-related diseases share several pathological commonalities, and emerging evidence suggests that targeting these biological processes ameliorates multiple age-related diseases. Immune aging plays a critical role in the pathogenesis of age-related diseases, given its involvement not only in controlling infection and cancer but also in facilitating tissue homeostasis and repair. Aging causes compositional and functional changes in both innate and adaptive immune cells, thereby significantly contributing to the pathogenesis of age-related disease and systemic low-grade inflammation, termed "inflammaging." This review article aims to describe the current understanding of immune aging and its impact on age-related diseases with particular emphasis on kidney and autoimmune diseases. In addition, this review highlights tertiary lymphoid structures (TLS) as a hallmark of immune aging, exploring their roles in inflammation, tissue damage, and potential therapeutic targeting.
Collapse
Affiliation(s)
- Yuki Sato
- Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, United States
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, United States
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
205
|
Chiang YH, Emmrich S, Vannini N. Metabolic Alterations in HSCs during Aging and Leukemogenesis. Physiology (Bethesda) 2025; 40:0. [PMID: 40019828 DOI: 10.1152/physiol.00054.2024] [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: 11/20/2024] [Revised: 12/15/2024] [Accepted: 02/23/2025] [Indexed: 04/26/2025] Open
Abstract
Aging is a multifaceted process associated with a functional decline in cellular function over time, affecting all lifeforms. During the aging process, metabolism, a fundamental hallmark of life (1), is profoundly altered. In the context of hematopoiesis, the proper function of hematopoietic stem cells, at the apex of the blood system, is tightly linked to their energy metabolism, which in turn shapes hematopoietic output. Here, we review the latest developments in our understanding of the metabolic states and changes in aged hematopoietic stem cells, molecular players and pathways involved in aged hematopoietic stem cell metabolism, the consequences of perturbed metabolism on clonal hematopoiesis and leukemogenesis, and pharmacologic/genetic strategies to reverse or rejuvenate altered metabolic phenotypes.
Collapse
Affiliation(s)
- Yi-Hsuan Chiang
- Department of Oncology, Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
| | - Stephan Emmrich
- Department of Oncology, Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
| | - Nicola Vannini
- Department of Oncology, Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
| |
Collapse
|
206
|
Li S, Liu Z, Chen Y, Feng S, Chen H, Zhao Y, He Y, Wang Q. Repaglinide platinum(IV) conjugates: Enhancing p53 signaling for antitumor and antimetastatic efficacy. J Inorg Biochem 2025; 268:112910. [PMID: 40199143 DOI: 10.1016/j.jinorgbio.2025.112910] [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: 01/04/2025] [Revised: 04/01/2025] [Accepted: 04/02/2025] [Indexed: 04/10/2025]
Abstract
The tumor suppressor p53 plays multiple roles at the crossroads of suppressing tumor development and metastasis. Here, a series of Repaglinide platinum(IV) conjugates promoting the p53 pathway were designed and prepared, which displayed potent antiproliferative and antimetastatic activities both in vitro and in vivo. Mechanistically, the expression of p53 was upregulated by the synergistic functions of the platinum core through causing severe DNA damage, and the RPG ligand via stimulating the lumican/p53/p21 pathway. The mitochondria-mediated apoptosis was initiated, involving the Bcl-2/Bax/caspase pathway. Pro-death autophagy was initiated with the upregulation of LC3II and down regulation of p62. Additionally, angiogenesis was suppressed by reversing tumor inflammation through the inhibition of key enzymes COX-2, MMP9, and VEGFA. Furthermore, antitumor immunity was enhanced by blocking the immune checkpoint PD-L1, which led to an increased presence of CD3+ and CD8+ T-cells within the tumor microenvironment.
Collapse
Affiliation(s)
- Suying Li
- Institute of Biopharmaceutical Research, State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, Shandong Key Laboratory of Applied Technology for Protein and Peptide Drugs, Liaocheng University, Liaocheng 252059, PR China
| | - Zhifang Liu
- Institute of Biopharmaceutical Research, State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, Shandong Key Laboratory of Applied Technology for Protein and Peptide Drugs, Liaocheng University, Liaocheng 252059, PR China
| | - Yan Chen
- Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, PR China
| | - Shuaiqi Feng
- Institute of Biopharmaceutical Research, State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, Shandong Key Laboratory of Applied Technology for Protein and Peptide Drugs, Liaocheng University, Liaocheng 252059, PR China
| | - Hengye Chen
- Institute of Biopharmaceutical Research, State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, Shandong Key Laboratory of Applied Technology for Protein and Peptide Drugs, Liaocheng University, Liaocheng 252059, PR China
| | - Yanna Zhao
- Institute of Biopharmaceutical Research, State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, Shandong Key Laboratory of Applied Technology for Protein and Peptide Drugs, Liaocheng University, Liaocheng 252059, PR China
| | - Yanqin He
- Institute of Biopharmaceutical Research, State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, Shandong Key Laboratory of Applied Technology for Protein and Peptide Drugs, Liaocheng University, Liaocheng 252059, PR China
| | - Qingpeng Wang
- Institute of Biopharmaceutical Research, State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, Shandong Key Laboratory of Applied Technology for Protein and Peptide Drugs, Liaocheng University, Liaocheng 252059, PR China.
| |
Collapse
|
207
|
Bashir B, Sethi P, Panda S, Manikyam HK, Vishwas S, Singh SK, Singh K, Jain D, Chaitanya MVNL, Coutinho HDM. Unravelling the epigenetic based mechanism in discovery of anticancer phytomedicine: Evidence based studies. Cell Signal 2025; 131:111743. [PMID: 40107479 DOI: 10.1016/j.cellsig.2025.111743] [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: 01/15/2025] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025]
Abstract
Epigenetic mechanisms play a crucial role in the normal development and maintenance of tissue-specific gene expression patterns in mammals. Disruption of these processes can result in changes to gene function and the transformation of cells into a malignant state. Cancer is characterized by widespread alterations in the epigenetic landscape, revealing that it involves not only genetic mutations but also epigenetic abnormalities. Recent progress in the field of cancer epigenetics has demonstrated significant reprogramming of various components of the epigenetic machinery in cancer, such as DNA methylation, modifications to histones, positioning of nucleosomes, and the expression of non-coding RNAs, particularly microRNAs. The ability to reverse epigenetic abnormalities has given rise to the hopeful field of epigenetic therapy, which has shown advancement with the recent approval by the FDA of three drugs targeting epigenetic mechanisms for the treatment of cancer. In the present manuscript, a comprehensive review has been presented about the role of understanding the epigenetic link between cancer and mechanisms by which phytomedicine offers treatment avenues. Further, this review deciphers the significance of natural products in the identification of epigenetic therapeutics, the diversity of their molecular targets, the use of nanotechnology, and the creation of new strategies for overcoming the inherent clinical challenges associated with developing these drug leads.
Collapse
Affiliation(s)
- Bushra Bashir
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144402, India
| | - Pranshul Sethi
- Department of Pharmacology, College of Pharmacy, Shri Venkateshwara University, Gajraula, Uttar Pradesh, India
| | - Satyajit Panda
- Department of Pharmaceutics, Institute of Pharmacy and Technology, Salipur, Cuttack, Odisha 754202, India
| | - Hemanth Kumar Manikyam
- Department of Chemistry, Faculty of science, North East Frontier Technical University, Arunachal Pradesh 791001, India
| | - Sukriti Vishwas
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144402, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144402, India
| | - Kuldeep Singh
- Department of Pharmacology, Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India.
| | - Divya Jain
- Department of Microbiology, School of Applied and Life sciences, Uttaranchal University, Dehradun, Uttarakhand 248007, India.
| | - M V N L Chaitanya
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144402, India.
| | | |
Collapse
|
208
|
Nakada T, Koga M, Takeuchi H, Doi K, Sugiyama H, Sakurai H. PP2A adapter protein IER5 induces dephosphorylation and degradation of MDM2, thereby stabilizing p53. Cell Signal 2025; 131:111739. [PMID: 40081547 DOI: 10.1016/j.cellsig.2025.111739] [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: 11/25/2024] [Revised: 02/11/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
The tumor suppressor p53 activates transcription of the IER5 gene, which encodes an adapter protein of protein phosphatase PP2A. IER5 binds to both the B55 regulatory subunit of PP2A and PP2A's target proteins, facilitating PP2A/B55-catalyzed dephosphorylation of these proteins. Here, we show that IER5 functions as a positive regulator of p53 by inhibiting its ubiquitination, thereby increasing cellular p53 levels. Mechanistically, this effect of IER5 requires its nuclear localization and binding to both PP2A/B55 and the p53 ubiquitin E3 ligase MDM2. Importantly, IER5 fails to inhibit p53 ubiquitination in cells treated with the MDM2 inhibitor Nutlin-3. The IER5-PP2A/B55 complex dephosphorylates MDM2 at Ser166, leading to MDM2 ubiquitination and a reduction in nuclear MDM2. Altogether, our data provide evidence that IER5-PP2A/B55 regulates the nuclear balance between MDM2 and p53 via MDM2 dephosphorylation.
Collapse
Affiliation(s)
- Taisei Nakada
- Division of Health Sciences, Graduate School of Medical Science, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa, Ishikawa 920-0942, Japan
| | - Mayuko Koga
- Division of Health Sciences, Graduate School of Medical Science, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa, Ishikawa 920-0942, Japan
| | - Hiroto Takeuchi
- Division of Health Sciences, Graduate School of Medical Science, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa, Ishikawa 920-0942, Japan
| | - Kuriko Doi
- Division of Health Sciences, Graduate School of Medical Science, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa, Ishikawa 920-0942, Japan
| | - Haruka Sugiyama
- Division of Health Sciences, Graduate School of Medical Science, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa, Ishikawa 920-0942, Japan
| | - Hiroshi Sakurai
- Division of Health Sciences, Graduate School of Medical Science, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa, Ishikawa 920-0942, Japan.
| |
Collapse
|
209
|
Wen M, Qiu P, Meng J, Zhao W, Wang X, Niu S, Tao C, Yu N, Chen Z, Xie D. Multifunctional nanozymes for sonodynamic-enhanced immune checkpoint blockade therapy by inactivating PI3K/AKT signal pathway. Biomaterials 2025; 318:123125. [PMID: 39893781 DOI: 10.1016/j.biomaterials.2025.123125] [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: 09/14/2024] [Revised: 01/09/2025] [Accepted: 01/23/2025] [Indexed: 02/04/2025]
Abstract
Insufficient activation efficacy and tumor immunosuppressive microenvironments hinder the infiltration of cytotoxic T lymphocytes (CTLs) for effective immunotherapy. Herein, the pH-selective multienzyme-mimetic nanozymes have been developed based on Pd-hemoporfin (Pd0/Pd2+‒H) nanoagents for tumor sono-immunotherapy via the phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) pathway inactivation. The Pd0/Pd2+‒H is capable of catalase-mimetic, peroxidase-mimetic, and sonodynamic effects, creating an O2-rich environment and elevating the reactive oxygen species (ROS) levels. The elevated ROS levels down-regulate the expression of PI3K and p-AKT on both gene and protein levels, leading to PI3K/AKT pathway inactivation. Subsequently, the augmented immunogenic cell death effectively recruits dendritic cells, presents tumor-associated antigens, and activates antitumor T-cell immunity. As a result, the combination of Pd0/Pd2+‒H and anti-programmed cell death protein ligand 1 results in growth restraints of primary and precaution of tumor metastases. This work offers insights into developing multienzyme-mimetic nanozymes in signaling pathway regulation and antitumor strategy.
Collapse
Affiliation(s)
- Mei Wen
- State Key Laboratory of Advanced Fiber Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Pu Qiu
- State Key Laboratory of Advanced Fiber Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Jialan Meng
- Department of Ultrasound, Songjiang Maternity & Child Health Hospital of Shanghai, Shanghai 201600, China
| | - Wenjing Zhao
- State Key Laboratory of Advanced Fiber Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Xiao Wang
- State Key Laboratory of Advanced Fiber Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Shining Niu
- State Key Laboratory of Advanced Fiber Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Cheng Tao
- State Key Laboratory of Advanced Fiber Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Nuo Yu
- State Key Laboratory of Advanced Fiber Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China.
| | - Zhigang Chen
- State Key Laboratory of Advanced Fiber Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China.
| | - Dong Xie
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China.
| |
Collapse
|
210
|
Liu X, Zhang L, Chen J, Shao W. Decoding intricate interactions between m6A modification with mRNAs and non-coding RNAs in cervical cancer: Molecular mechanisms and clinical implications. Cell Signal 2025; 131:111745. [PMID: 40107480 DOI: 10.1016/j.cellsig.2025.111745] [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: 10/10/2024] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 03/22/2025]
Abstract
N6-methyladenosine (m6A) methylation is the most prevalent RNA modification that is regulated by three regulatory factors: "writers", "erasers" and "readers". m6A modification regulates RNA stability and other mechanisms, including translation, cleavage, and degradation. Current research has demonstrated that m6A methylation is involved in the regulation of occurrence and development of cancers by controlling the expression of cancer-related genes. This review summarizes the role of m6A modification on messenger RNAs (mRNAs) and non-coding RNAs (ncRNAs) in cervical cancer (CC). We highlight the dual role of m6A regulatory factors, which act as oncogenes or tumor suppressors depending on the cellular context and downstream targets. Additionally, we examine how ncRNAs reciprocally regulate m6A modification in two ways: by guiding the deposition or removal of m6A modifications on RNA targets, and by modulating the expression of m6A regulatory factors. These interactions further contribute to tumor progression. Furthermore, the therapeutic potential of targeting m6A modification has been emphasized in CC. Moreover, recent advances in small-molecule inhibitors targeting m6A regulators and RNA-based therapies which may offer new treatment strategies have been summarized. Finally, we discuss the current challenges in m6A modification research and provide suggestions for future research directions. This review aims to deepen the understanding of m6A modification in CC and contribute to the development of targeted and personalized treatment strategies.
Collapse
Affiliation(s)
- Xuefei Liu
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, Anhui, China; First Clinical Medical College, Anhui Medical University, Hefei, Anhui, China
| | - Lizhi Zhang
- First Clinical Medical College, Anhui Medical University, Hefei, Anhui, China
| | - Ji Chen
- Department of Obstetrics, The Third Affiliated Hospital of Anhui Medical University, Hefei 230061, Anhui, China
| | - Wei Shao
- Department of Microbiology and Parasitology, Anhui Provincial Laboratory of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, Anhui, China.
| |
Collapse
|
211
|
Ma C, Wang J, Li Q, Wu Y, Yu Z, Chao Y, Liu Z, Chen G. Injectable oxidized high-amylose starch hydrogel scaffold for macrophage-mediated glioblastoma therapy. Biomaterials 2025; 318:123128. [PMID: 39884130 DOI: 10.1016/j.biomaterials.2025.123128] [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: 11/07/2024] [Revised: 01/19/2025] [Accepted: 01/23/2025] [Indexed: 02/01/2025]
Abstract
Glioblastoma, characterized by rapid proliferation and invasiveness, is largely resistant to current treatment modalities. A major obstacle is the blood-brain barrier (BBB), which restricts the delivery of therapeutic agents as well as the infiltration of effective immune cells into glioblastoma. In this study, we developed an injectable oxidized high-amylose starch hydrogel (OHASM) to serve as a biomaterial scaffold for the delivery of macrophages and macrophage-polarizing drugs, aiming to bypass the BBB and enhance glioblastoma treatment. The in vitro and in vivo experiments confirmed the efficacy of the hydrogel in loading and delivering macrophages and polarizing drugs against glioblastoma. Additionally, the hydrogel's interconnected porous structure was conducive to cellular growth and activity, and its slow release of therapeutics contributed to the extended survival of treated mice in a mouse GL261 glioblastoma tumor model. The immunological mechanisms underlying the therapeutic efficacy were further elucidated, revealing the potential of the hydrogel system to modulate macrophage polarization and induce apoptosis in tumor cells via the poly ADP-ribose polymerase (PARP) pathway. The study underscores the potential of the hydrogel-based macrophage delivery strategy as an effective and safe treatment for glioblastoma, offering a promising avenue for clinical management of this aggressive brain cancer.
Collapse
Affiliation(s)
- Cheng Ma
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215006 China
| | - Jiahe Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215006 China
| | - Qiaofeng Li
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123 China
| | - Yuzhe Wu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123 China
| | - Zhengquan Yu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215006 China
| | - Yu Chao
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123 China.
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123 China.
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215006 China.
| |
Collapse
|
212
|
Xiong S, Zhang Y, Zhou X, Pant V, Mirani A, Gencel-Augusto J, Chau G, You MJ, Lozano G. Dependence on Mdm2 for Mdm4 inhibition of p53 activity. Cancer Lett 2025; 621:217622. [PMID: 40081463 DOI: 10.1016/j.canlet.2025.217622] [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: 01/27/2025] [Revised: 03/06/2025] [Accepted: 03/09/2025] [Indexed: 03/16/2025]
Abstract
Both Mdm2 and Mdm4 inhibit p53 activity by masking of its transcriptional activation domain. In addition, Mdm2 functions as an E3 ubiquitin ligase, targeting p53 for degradation. The amino terminus of Mdm4 binds wild type and mutant p53 while its RING domain, which lacks E3 ligase activity, is required for heterodimerization with Mdm2. To determine how these domains of Mdm4 regulate p53, we generated mouse models with either a deletion of the Mdm4 RING domain (Mdm4ΔR) or all of Mdm4 (Mdm4─) on a hypomorphic (p53neo) background. Mdm4ΔR mice exhibited elevated p53 levels and activity, albeit to a lesser extent than mice with complete Mdm4 loss, indicating that the amino terminus of Mdm4 contributes to p53 inhibition. Moreover, in the absence of Mdm2, neither the deletion of the Mdm4 RING domain nor the complete loss of Mdm4 further increased p53 protein levels on a mutant p53 background, indicating that Mdm4 modulates Mdm2 in its regulation of p53 stability. Collectively, our findings suggest that Mdm4 contributes to p53 inhibition by modulating Mdm2 activity via both its amino terminus and RING domains.
Collapse
Affiliation(s)
- Shunbin Xiong
- Department of Genetics, UT M.D. Anderson Cancer Center, Houston, TX, USA
| | - Yun Zhang
- Department of Pharmaceutical Sciences, Joan M. Lafleur College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, 77004, USA
| | - Xin Zhou
- Department of Pediatrics, UT M.D. Anderson Cancer Center, Houston, TX, USA
| | - Vinod Pant
- Department of Genetics, UT M.D. Anderson Cancer Center, Houston, TX, USA
| | - Akshita Mirani
- Department of Genetics, UT M.D. Anderson Cancer Center, Houston, TX, USA
| | | | - Gilda Chau
- Department of Genetics, UT M.D. Anderson Cancer Center, Houston, TX, USA
| | - M James You
- Department of Hematopathology, UT M.D. Anderson Cancer Center, Houston, TX, USA
| | - Guillermina Lozano
- Department of Genetics, UT M.D. Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
213
|
Feng Y, Qiu H, Chen D. Regulation of Stem Cell Function by NAD . Physiology (Bethesda) 2025; 40:0. [PMID: 39907078 DOI: 10.1152/physiol.00052.2024] [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: 11/11/2024] [Revised: 01/20/2025] [Accepted: 01/28/2025] [Indexed: 02/06/2025] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+), a coenzyme in cellular metabolism, has never ceased to capture the fascination of scientists since its discovery in 1906. The expansion of NAD+'s function from cellular metabolism to DNA repair, gene regulation, cell signaling, and aging reflects the central role of cellular metabolism in orchestrating the diverse cellular pathways. In the past decade, NAD+ has emerged as a key regulator of stem cells, opening the door to potential approaches for regenerative medicine. Here we reflect on how the field of NAD+ regulation of stem cells has evolved since a decade ago, when sirtuins, NAD+-dependent enzymes, were shown to be critical regulators of stem cells. We review the recent development on how NAD+ is regulated in stem cells to influence fate decision. We discuss the difference in NAD+ regulation of normal and cancer stem cells. Finally, we consider the consequences of NAD+ regulation of stem cells for health and diseases.
Collapse
Affiliation(s)
- Yufan Feng
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, California, United States
| | - Huixian Qiu
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, California, United States
| | - Danica Chen
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, California, United States
| |
Collapse
|
214
|
Huang Y, Chen L, Chen Y, Zhou S, Xie X, Xie J, Yu M, Chen J. High-density lipoprotein-based nanoplatform reprograms tumor microenvironment and enhances chemotherapy against pancreatic ductal adenocarcinoma. Biomaterials 2025; 318:123147. [PMID: 39908877 DOI: 10.1016/j.biomaterials.2025.123147] [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: 10/16/2024] [Revised: 11/25/2024] [Accepted: 01/26/2025] [Indexed: 02/07/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is highly aggressive, with limited success in traditional therapies due to the fibrotic, immunosuppressive, pro-metastatic tumor microenvironment (TME), which collectively impede the drug accumulation and accelerate the tumor progression. In this work, we developed a PDAC-customized nutrient-mimicking reconstituted high-density lipoprotein (rHDL) capable of efficiently co-encapsulate versatile TME regulating cannabidiol and cytotoxic gemcitabine to simultaneously reprogram TME while suppressing PDAC progression. Specifically, a small-sized, nutrient-like rHDL was constructed to realize deep PDAC parenchyma penetration and efficient intra-tumoral uptake. Next, natural herbal compound cannabidiol was screened and incorporated into rHDL to regulate TME via attenuating fibrosis, reliving immunosuppression and mitigating metastatic tendency. At last, gemcitabine, the PDAC gold standard first-line therapy was co-delivered by the PDAC-customized rHDL to overcome drug resistance and amplify its PDAC suppression. Our findings demonstrate the feasibility of an integrated multi-stage TME regulation strategy for improved PDAC therapy, and might represent a modality in promoting chemotherapy against PDAC.
Collapse
Affiliation(s)
- Yukun Huang
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China; Department of Pharmacology and Chemical Biology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Liang Chen
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China
| | - Yu Chen
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China
| | - Songlei Zhou
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China
| | - Xiaoying Xie
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China
| | - Jing Xie
- Department of Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Minghua Yu
- Fudan University Clinical Research Center for Cell-based Immunotherapy & Department of Oncology, Fudan University Pudong Medical Center, 2800 Gongwei Road, Shanghai, 201399, China
| | - Jun Chen
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China.
| |
Collapse
|
215
|
Chen L, Li Y, Zhao H, Huang J, Yan H, Lin X, Zhao B. Pan-cancer analysis of MET mutation and its association with the efficacy of immune checkpoint blockade. Genes Dis 2025; 12:101450. [PMID: 40330151 PMCID: PMC12053711 DOI: 10.1016/j.gendis.2024.101450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 08/15/2024] [Accepted: 08/25/2024] [Indexed: 05/08/2025] Open
Abstract
The mesenchymal-epithelial transition factor (MET) proto-oncogene plays important roles during tumor development. Recently, evidence has revealed MET signaling may impact tumor immunogenicity and regulate the immune response. Here we conducted a comprehensive bioinformatic and clinical analysis to explore the characteristics of MET mutation and its association with the outcomes in pan-cancer immunotherapy. In 4149 patients with 12 tumor types treated with immune checkpoint inhibitors, MET mutation indicated favorable overall survival (hazard ratio = 0.61; 95% CI, 0.50-0.74; P < 0.001), progression-free survival (hazard ratio = 0.74; 95% CI, 0.60-0.92; P = 0.01), and objective response rate (40.3% vs. 28.1%; P = 0.003). Moreover, we developed a nomogram to estimate the 12-month and 24-month survival probabilities after the initiation of immunotherapy. Further multi-omics analysis on both intrinsic and extrinsic immune landscapes revealed that MET mutation enhanced tumor immunogenicity, enriched infiltration of immune cells, and improved immune responses. In summary, MET mutation improves cancer immunity and is an independent biomarker for favorable outcomes in pan-cancer immunotherapy. These results may influence clinical practices, guide treatment decision-making, and develop immunotherapy for personalized care.
Collapse
Affiliation(s)
- Lijin Chen
- Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China
- Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, Fujian 362000, China
| | - Yingying Li
- Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, Fujian 362000, China
| | - Hong Zhao
- The Cancer Center of The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, Guangdong 519000, China
| | - Jinyuan Huang
- Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, Fujian 362000, China
| | - Huimeng Yan
- Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, Fujian 362000, China
| | - Xiaoyan Lin
- Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, China
| | - Bin Zhao
- Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, Fujian 362000, China
| |
Collapse
|
216
|
Liu H, Xue H, Guo Q, Xue X, Yang L, Zhao K, Liu Y. Ferroptosis meets inflammation: A new frontier in cancer therapy. Cancer Lett 2025; 620:217696. [PMID: 40189012 DOI: 10.1016/j.canlet.2025.217696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 03/26/2025] [Accepted: 04/03/2025] [Indexed: 04/10/2025]
Abstract
Ferroptosis, an iron-dependent form of regulated cell death driven by lipid peroxidation, has emerged as a critical player in cancer pathogenesis. Concurrently, inflammation, a key biological response to tissue injury or infection, significantly influences cancer development and progression. The interplay between ferroptosis and inflammation represents a promising yet underexplored area of research. This review synthesizes recent advances in understanding the molecular mechanisms governing their interaction, emphasizing how ferroptosis triggers inflammatory responses and how inflammatory mediators, such as TNF-α, regulate ferroptosis through iron metabolism and lipid peroxidation pathways. Key molecular targets within the ferroptosis-inflammation axis, including GPX4, ACSL4, and the NF-κB signaling pathway, offer therapeutic potential for cancer treatment. By modulating these targets, it may be possible to enhance ferroptosis and fine-tune inflammatory responses, thereby improving therapeutic outcomes. Additionally, this review explores the broader implications of targeting the ferroptosis-inflammation interplay in disease treatment, highlighting opportunities for developing innovative strategies to combat cancer. By bridging the gap in current knowledge, this review provides a comprehensive resource for researchers and clinicians, offering insights into the therapeutic potential of this intricate biological relationship.
Collapse
Affiliation(s)
- Hu Liu
- Department of Oncology Surgery, Shanghai Mengchao Hospital, Shanghai University, Shanghai, 202800, China
| | - Hui Xue
- Department of Oncology Surgery, Shanghai Mengchao Hospital, Shanghai University, Shanghai, 202800, China
| | - Qian Guo
- Department of Rhinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xutong Xue
- Boston Children's Hospital, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Lixue Yang
- Department of Oncology Surgery, Shanghai Mengchao Hospital, Shanghai University, Shanghai, 202800, China.
| | - Kaijun Zhao
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| | - Yu'e Liu
- Boston Children's Hospital, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA; Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| |
Collapse
|
217
|
Wang Y, Yu Y, Yu J, Wang C, Wang Y, Fu R, Zhang C. The intersections between neuroscience and medulloblastoma. Cancer Lett 2025; 620:217660. [PMID: 40154912 DOI: 10.1016/j.canlet.2025.217660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/17/2025] [Accepted: 03/19/2025] [Indexed: 04/01/2025]
Abstract
Medulloblastoma (MB) represents the most common malignant central nervous system tumor in childhood. The nervous system plays a critical role in the progression of MB, with interactions between the nervous system and cancer significantly influencing oncogenesis, tumor growth, invasion, stemness, and metabolism. These interactions also regulate angiogenesis, metastatic dissemination, the tumor immune microenvironment, and drug resistance. Investigating the nervous system-MB axis holds promise for identifying diagnostic markers, prognostic biomarkers, and therapeutic targets. It also provides insights into the molecular mechanisms underlying MB and informs the development of novel therapeutic strategies. This review summarizes the latest advancements in understanding the interplay between the nervous system and MB, including the role of glial cells in MB and the potential of drug repurposing targeting nervous system components for MB treatment. These findings underscore promising diagnostic and therapeutic opportunities for MB management. Additionally, we outline future research directions in neurosciences that may pave the way for innovative therapeutic approaches and deepen our understanding of this complex disease.
Collapse
Affiliation(s)
- Yafei Wang
- Department of Pediatric Neurosurgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Ying Yu
- Department of Pediatric Neurosurgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Jiahua Yu
- Department of Pediatric Neurosurgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Cheng Wang
- Department of Pediatric Neurosurgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China; Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yunkun Wang
- Department of Pediatric Neurosurgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Runxi Fu
- Department of Pediatric Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China; Shanghai Institute for Pediatric Research, Shanghai, China
| | - Chenran Zhang
- Department of Pediatric Neurosurgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
218
|
Kundu M, Greer YE, Lobanov A, Ridnour L, Donahue RN, Ng Y, Ratnayake S, White K, Voeller D, Weltz S, Chen Q, Lockett SJ, Cam M, Meerzaman D, Wink DA, Weigert R, Lipkowitz S. TRAIL induces cytokine production via the NFkB2 pathway promoting neutrophil chemotaxis and neutrophil-mediated immune-suppression in triple negative breast cancer cells. Cancer Lett 2025; 620:217692. [PMID: 40187604 PMCID: PMC12049148 DOI: 10.1016/j.canlet.2025.217692] [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: 09/06/2024] [Revised: 03/31/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a potential cancer therapeutic that induces apoptosis in cancer cells while sparing the non-malignant cells in preclinical models. However, its efficacy in clinical trials has been limited, suggesting unknown mechanisms modulating TRAIL activity in patients. We hypothesized that TRAIL treatment elicits transcriptional changes in triple negative breast cancer (TNBC) cells that alter the immune milieu. RNAseq analysis of MDA-MB-231 cells along with validation in additional cell lines demonstrated that TRAIL induced cytokines such as CXCLs 1, 2, 3, 8,11 and IL-6, which are known to modify neutrophil function. Mechanistically, TRAIL dependent induction of the cytokines was predominantly mediated by death receptor 5, caspase-8 and the non-canonical NFKB2 pathway. These cytokines produced by TRAIL-treated TNBC cells enhanced chemotaxis of normal human donor isolated neutrophils. Using TNBC xenograft models, TRAIL induced activation of NFkB2 pathway, cytokine production and increased neutrophil recruitment into the tumors. Moreover, preincubation of neutrophils in supernatants from TRAIL-treated TNBC cells significantly impaired neutrophil function as measured by reduced respiratory burst and cytotoxic effect against TNBC cells. Transcriptomic analysis of neutrophils incubated with either TRAIL alone or supernatant of TRAIL-treated TNBC cells revealed increased expression of inflammatory cytokines, immune modulatory genes, immune checkpoint genes, and genes implicated in delayed neutrophil apoptosis. Functional studies showed that these neutrophils suppress T cell proliferation and augment Treg suppressive phenotype. Collectively, our study demonstrates a novel role of TRAIL-induced NFKB2-dependent cytokine production that promotes neutrophil chemotaxis and neutrophil-mediated immune suppression.
Collapse
Affiliation(s)
- Manjari Kundu
- Women's Malignancies Branch, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Yoshimi E Greer
- Women's Malignancies Branch, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Alexei Lobanov
- Center for Cancer Research Collaborative Bioinformatics Resource (CCBR), NCI, NIH, Bethesda, MD, USA
| | - Lisa Ridnour
- Cancer Innovation Laboratory, Center for Cancer Research (CCR), NCI, NIH, Frederick, MD, USA
| | - Renee N Donahue
- Center for Immuno-Oncology, CCR, NCI, NIH, Bethesda, MD, USA
| | - Yeap Ng
- Laboratory of Cellular and Molecular Biology, CCR, NCI, NIH, Bethesda, MD, USA
| | - Shashi Ratnayake
- Computational Genomics and Bioinformatics Branch, Center for Biomedical Informatics and Information Technology (CBIIT), NCI, NIH, Rockville, MD, USA
| | - Karley White
- Women's Malignancies Branch, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Donna Voeller
- Women's Malignancies Branch, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Sarah Weltz
- Women's Malignancies Branch, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Qingrong Chen
- Computational Genomics and Bioinformatics Branch, Center for Biomedical Informatics and Information Technology (CBIIT), NCI, NIH, Rockville, MD, USA
| | - Stephen J Lockett
- Optical Microscopy and Analysis Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Maggie Cam
- Center for Cancer Research Collaborative Bioinformatics Resource (CCBR), NCI, NIH, Bethesda, MD, USA
| | - Daoud Meerzaman
- Computational Genomics and Bioinformatics Branch, Center for Biomedical Informatics and Information Technology (CBIIT), NCI, NIH, Rockville, MD, USA
| | - David A Wink
- Cancer Innovation Laboratory, Center for Cancer Research (CCR), NCI, NIH, Frederick, MD, USA
| | - Roberto Weigert
- Laboratory of Cellular and Molecular Biology, CCR, NCI, NIH, Bethesda, MD, USA
| | - Stanley Lipkowitz
- Women's Malignancies Branch, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA.
| |
Collapse
|
219
|
Maleszewska M, Roura AJ, Dabrowski MJ, Draminski M, Wojtas B. Decoding glioblastoma's diversity: Are neurons part of the game? Cancer Lett 2025; 620:217666. [PMID: 40147584 DOI: 10.1016/j.canlet.2025.217666] [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: 01/04/2025] [Revised: 03/18/2025] [Accepted: 03/21/2025] [Indexed: 03/29/2025]
Abstract
Glioblastoma multiforme (GBM, WHO Grade 4) is a highly aggressive primary brain tumor with limited treatment options and a poor prognosis. A key challenge in GBM therapy lies in its pronounced heterogeneity, both within individual tumors (intratumoral) and between patients (intertumoral). Historically, neurons have been underexplored in GBM research; however, recent studies reveal that GBM development is closely linked to neural and glial progenitors, often mimicking neurodevelopmental processes in a dysregulated manner. Beyond damaging neuronal tissue, GBM actively engages with neurons to promote pro-tumorigenic signaling, including neuronal hyperexcitability and seizures. Single-cell RNA sequencing (scRNA-seq) has revolutionized our understanding of the tumor microenvironment (TME), uncovering the critical roles of immune cells, endothelial cells, and astrocytes in tumor progression. However, technical limitations of scRNA-seq hinder its ability to capture the transcriptomes of neurons, necessitating the use of single-nucleus RNA sequencing (snRNA-seq) to study these interactions at single-cell resolution. This work collects the emerging insights of glioblastoma-neuron interactions, focusing on how GBM exploits neurodevelopmental pathways and reshapes neuronal networks. Moreover, we perform bioinformatic analysis of publicly available snRNA-seq datasets to propose putative cell-cell interactions driving glioma-neuronal dynamics. This study delineates key signaling pathways and underscores the need for further investigation to evaluate their potential as therapeutic targets.
Collapse
Affiliation(s)
- Marta Maleszewska
- Department of Animal Physiology, Institute of Experimental Zoology, Faculty of Biology, University of Warsaw, 1 Miecznikowa Str, 02-096, Warsaw, Poland.
| | - Adrià-Jaume Roura
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Barcelona, Spain
| | - Michal J Dabrowski
- Computational Biology Group, Institute of Computer Science of the Polish Academy of Sciences, Warsaw, Poland
| | - Michal Draminski
- Computational Biology Group, Institute of Computer Science of the Polish Academy of Sciences, Warsaw, Poland
| | - Bartosz Wojtas
- Laboratory of Sequencing, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
220
|
Li J, Xu S, Zhan Y, Lv X, Sun Z, Man L, Yang D, Sun Y, Ding S. CircRUNX1 enhances the Warburg effect and immune evasion in non-small cell lung cancer through the miR-145/HK2 pathway. Cancer Lett 2025; 620:217639. [PMID: 40090573 DOI: 10.1016/j.canlet.2025.217639] [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: 11/19/2024] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 03/18/2025]
Abstract
Non-small cell lung cancer (NSCLC) is acknowledged as the primary subtype of lung cancer. The Warburg effect, marked by elevated glucose consumption and lactate fermentation, is a prevalent characteristic of NSCLC. The mechanisms by which circRNA mediates the regulation of the Warburg effect and immune evasion in NSCLC remain unclear. This study found an elevated circRNA, circRUNX1, whiche promotes glycolysis and lactate generation, resulting in the infiltration of regulatory T cell (Treg) in NSCLC. circRUNX1 acts as a miR-145 sponge, inhibiting its negative regulation of the target gene HK2, therefore facilitating glycolysis and lactate generation. The accumulation of lactic acid in the tumor microenvironment promotes Treg cell proliferation and aids immune evasion. Functionally, the suppression of circRUNX1 significantly impedes tumor development both in vitro and in vivo. These findings collectively clarity a previously unexamined mechanism linking the circRUNX1/miR-145/HK2 axis in regulation of the Warburg effect and immune evasion in NSCLC.
Collapse
MESH Headings
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Humans
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/metabolism
- Lung Neoplasms/genetics
- Lung Neoplasms/immunology
- Lung Neoplasms/pathology
- Lung Neoplasms/metabolism
- RNA, Circular/genetics
- RNA, Circular/metabolism
- Warburg Effect, Oncologic
- Animals
- Tumor Microenvironment/immunology
- Mice
- Hexokinase/genetics
- Hexokinase/metabolism
- Gene Expression Regulation, Neoplastic
- Cell Line, Tumor
- Immune Evasion
- Cell Proliferation
- Glycolysis
- T-Lymphocytes, Regulatory/immunology
- Signal Transduction
- Lactic Acid/metabolism
- Tumor Escape
- Female
Collapse
Affiliation(s)
- Jinyou Li
- Department of Thoracic Surgery, Affiliated Hospital of Jiangnan University, Wuxi, 214000, China; Department of Thoracic Surgery, Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Shiwei Xu
- Department of Thoracic Surgery, Affiliated Hospital of Jiangnan University, Wuxi, 214000, China; Department of Thoracic Surgery, Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Yangyang Zhan
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Navy Military Medical University, 225 Changhai Road, Yangpu District, Shanghai, China; Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| | - Xinyi Lv
- Department of Thoracic Surgery, Affiliated Hospital 2 of Nantong University, Nantong First People's Hospital, Nantong, 226001, China; School of Medicine, Nantong University, Nantong, 226001, China
| | - Zhenyu Sun
- Department of Thoracic Surgery, Affiliated Hospital of Jiangnan University, Wuxi, 214000, China; Department of Thoracic Surgery, Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Li Man
- Department of Medical Oncology, Anshan Cancer Hospital, Anshan, 114000, China
| | - Donghua Yang
- New York College of Traditional Chinese Medicine, 200 Old Country Rd, Suite 500, Mineola, NY, 11501, USA
| | - Yahong Sun
- Department of Respiratory and Critical Care Medicine, Haining People's Hospital, Haining, 314400, China.
| | - Shengguang Ding
- Department of Thoracic Surgery, Affiliated Hospital 2 of Nantong University, Nantong First People's Hospital, Nantong, 226001, China.
| |
Collapse
|
221
|
Haykal MM, Rodrigues-Ferreira S, El Botty R, Sourd L, Marangoni E, Varin M, Denis A, Nahmias C. Targeting WEE1 kinase as a therapeutic strategy in ATIP3-deficient breast cancers. Cancer Lett 2025; 620:217665. [PMID: 40127815 DOI: 10.1016/j.canlet.2025.217665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/04/2025] [Accepted: 03/21/2025] [Indexed: 03/26/2025]
Abstract
ATIP3-deficient breast cancers represent a subset of aggressive tumors with limited therapeutic options and poor prognosis. Here, we screened a panel of cell cycle kinase inhibitors to identify novel targets for these tumors. We show that loss of ATIP3 sensitizes breast cancer cells to WEE1 inhibition, resulting in aberrant mitoses characterized by detachment of centromere proteins from DNA and chromosome pulverization. This phenotype arises from excessive replication stress and DNA damage in S-phase, combined with premature mitotic entry driven by untimely CDK1 activation. Mechanistically, we identify DNA2 helicase/nuclease as a key mediator of chromosome pulverization. Importantly, the heightened sensitivity of ATIP3-deficient cells to WEE1 inhibition provides a strong rationale for clinical exploration of WEE1-targeted therapies. Furthermore, combining WEE1 and PKMYT1 inhibitors enhances therapeutic efficacy, offering a promising strategy for personalized treatment in ATIP3-deficient breast cancers.
Collapse
Affiliation(s)
- Maria M Haykal
- Institut Gustave Roussy, Inserm U981, Biomarqueurs prédictifs et Nouvelles Stratégies Thérapeutiques en Oncologie, 94800, Villejuif, France; Université Paris-Saclay, 91400, Orsay, France
| | - Sylvie Rodrigues-Ferreira
- Institut Gustave Roussy, Inserm U981, Biomarqueurs prédictifs et Nouvelles Stratégies Thérapeutiques en Oncologie, 94800, Villejuif, France; Université Paris-Saclay, 91400, Orsay, France; Inovarion, 75005, Paris, France
| | - Rania El Botty
- Translational Research Department, Institut Curie, PSL University, 75005, Paris, France
| | - Laura Sourd
- Translational Research Department, Institut Curie, PSL University, 75005, Paris, France
| | - Elisabetta Marangoni
- Translational Research Department, Institut Curie, PSL University, 75005, Paris, France
| | | | | | - Clara Nahmias
- Institut Gustave Roussy, Inserm U981, Biomarqueurs prédictifs et Nouvelles Stratégies Thérapeutiques en Oncologie, 94800, Villejuif, France; Université Paris-Saclay, 91400, Orsay, France.
| |
Collapse
|
222
|
Cheng C, Cui L, Cui X, Zhan Q, Ju J, Hong B, Huang Y, Ding Y, Xu H, Qiu T, Kang C, Liu X, Wang Q, Zeng L. ADAM12 promotes temozolomide resistance in glioblastoma by activating the TNF-α - NF-κB pathway. Cancer Lett 2025; 620:217684. [PMID: 40180114 DOI: 10.1016/j.canlet.2025.217684] [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: 10/05/2024] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 04/05/2025]
Abstract
Development of temozolomide (TMZ) resistance is a critical factor contributing to a poor prognosis in glioma patients. TMZ resistance is also closely associated with the phosphorylation level of NF-κB, yet targeted inhibition of NF-κB activity in glioma can be leveraged to overcome TMZ resistance. ADAM12, a protein significantly overexpressed in glioma cells, is implicated in the pathogenesis and progression of glioma, yet its role in the development of TMZ resistance is completely understood. We found that knockdown of ADAM12 was shown to arrest the glioma cell cycle, enhance apoptosis, inhibit DNA damage repair mechanisms, and sensitize glioma cells to TMZ. Targeting ADAM12 in vivo was found to increase the sensitivity of glioma cells to TMZ. Survival analysis indicated that ADAM12 serves as a prognostic marker for TMZ treatment. Using ELISA and protein interaction predictions via docking simulation, we identified the TNF-α shedding function of ADAM12 as a critical regulator of glioma progression. Furthermore, in glioma cell lines with unmethylated MGMT, the knockdown of ADAM12 enhanced sensitivity to TMZ by inhibiting the TNF-α/NF-κB pathway and reducing MGMT expression. In all, these results demonstrated that ADAM12 aids in shedding of membrane-bound TNF-a to drive TMZ resistance in glioma.
Collapse
Affiliation(s)
- Chunchao Cheng
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital and Key Laboratory of Neurotrauma, Variation, and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin, 300052, China; Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Longtao Cui
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital and Key Laboratory of Neurotrauma, Variation, and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin, 300052, China; Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xiaoteng Cui
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital and Key Laboratory of Neurotrauma, Variation, and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin, 300052, China; Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Qi Zhan
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital and Key Laboratory of Neurotrauma, Variation, and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin, 300052, China; Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Jiasheng Ju
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital and Key Laboratory of Neurotrauma, Variation, and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin, 300052, China; Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Biao Hong
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital and Key Laboratory of Neurotrauma, Variation, and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin, 300052, China; Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Yanping Huang
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital and Key Laboratory of Neurotrauma, Variation, and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin, 300052, China; Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Yaqing Ding
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital and Key Laboratory of Neurotrauma, Variation, and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin, 300052, China; Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Hanyi Xu
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital and Key Laboratory of Neurotrauma, Variation, and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin, 300052, China; Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Tian Qiu
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital and Key Laboratory of Neurotrauma, Variation, and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin, 300052, China; Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Chunsheng Kang
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital and Key Laboratory of Neurotrauma, Variation, and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin, 300052, China; Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xiaomin Liu
- Neuro-Oncology Center, Tianjin Huanhu Hospital, Tianjin, 300350, China.
| | - Qixue Wang
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital and Key Laboratory of Neurotrauma, Variation, and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin, 300052, China; Tianjin Medical University General Hospital, Tianjin, 300052, China.
| | - Liang Zeng
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| |
Collapse
|
223
|
Gerakopoulos V, Ramos C, Müller C, Walterskirchen N, Vintila S, Zotter C, Ilg M, Pap A, Riss S, Bergmann M, Unger LW, Vogt AB, Oehler R, Lukowski SW. Single-cell transcriptomic analysis identifies tissue-specific fibroblasts as the main modulators of myeloid cells in peritoneal metastasis of different origin. Cancer Lett 2025; 620:217678. [PMID: 40154914 DOI: 10.1016/j.canlet.2025.217678] [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: 03/12/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Colorectal cancer (CRC) peritoneal metastasis (CPM) is related to limited therapy options and poor prognosis. Although stromal cells heavily infiltrate most CPMs, interactions between different cell types in their microenvironment remain unclear. Here, we investigated tumor and distant normal tissue from CPM and CRC patients using single-cell RNA sequencing. Investigating the incoming and outgoing signals between cells revealed that fibroblasts dominate the CPM signaling landscape with myeloid cells as their strongest interaction partner. Using immunohistochemistry, we confirmed that fibroblasts co-localize with macrophages in the CPM microenvironment. A fibroblast sub-population detected only in CPM and normal peritoneum demonstrated immunoregulatory properties in co-culture experiments, and was further detected in additional peritoneal malignancies derived from ovarian and gastric origin. This novel fibroblast type and its communication with macrophages could be attractive targets for therapeutic interventions in CPM and potentially peritoneal surface malignancies in general.
Collapse
Affiliation(s)
- Vasileios Gerakopoulos
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Cristiano Ramos
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Catharina Müller
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Natalie Walterskirchen
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Stefania Vintila
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Chiara Zotter
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Mathias Ilg
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim RCV GmBH & Co KG., Dr. Boehringer Gasse 5-11, 1120, Vienna, Austria
| | - Anna Pap
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim RCV GmBH & Co KG., Dr. Boehringer Gasse 5-11, 1120, Vienna, Austria
| | - Stefan Riss
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Michael Bergmann
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Lukas W Unger
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria; Dept. of Colorectal Surgery, Oxford University Hospitals, Old Rd, Headington, Oxford, OX3 7LE, United Kingdom
| | - Anne B Vogt
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim RCV GmBH & Co KG., Dr. Boehringer Gasse 5-11, 1120, Vienna, Austria
| | - Rudolf Oehler
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, 1090, Vienna, Austria.
| | - Samuel W Lukowski
- Cancer Immunology and Immune Modulation, Boehringer Ingelheim RCV GmBH & Co KG., Dr. Boehringer Gasse 5-11, 1120, Vienna, Austria
| |
Collapse
|
224
|
Parimita S, Das A, Samanta S. Vestigial-like family member 1 (VGLL1): An emerging candidate in tumor progression. Biochem Biophys Res Commun 2025; 766:151889. [PMID: 40300335 DOI: 10.1016/j.bbrc.2025.151889] [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: 03/08/2025] [Revised: 04/19/2025] [Accepted: 04/23/2025] [Indexed: 05/01/2025]
Abstract
Vestigial-like family member 1 (VGLL1), a product of an X-linked gene (VGLL1), belongs to a family of transcriptional co-activators including VGLL2, VGLL3 and VGLL4. These proteins are called vestigial-like because of the structural and functional similarities with the Drosophila ortholog vestigial (vg). VGLL1 is usually expressed in human placenta, and has also been detected in many aggressive cancers. For this reason, it is called an onco-placental protein. It can bind and activate the TEA-domain containing transcription factors TEAD1-4, and the interaction is mediated through a conserved 'valine-x-x-histidine-phenylalanine' domain (VxxHF, x denotes any amino acid) present in VGLL1 protein. Prior studies indicate a pro-tumorigenic role for this protein in several cancers including carcinoma of the breast. This review aims at summarizing our present knowledge about the functions of VGLL1, and the mechanisms that regulate its expression in cancer.
Collapse
Affiliation(s)
- Shubhashree Parimita
- Department of Applied Biology, Council of Scientific & Industrial Research-Indian Institute of Chemical Technology (CSIR-IICT), Uppal Road, Tarnaka, Hyderabad, TS, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad, 201002, India
| | - Amitava Das
- Department of Applied Biology, Council of Scientific & Industrial Research-Indian Institute of Chemical Technology (CSIR-IICT), Uppal Road, Tarnaka, Hyderabad, TS, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad, 201002, India
| | - Sanjoy Samanta
- Department of Applied Biology, Council of Scientific & Industrial Research-Indian Institute of Chemical Technology (CSIR-IICT), Uppal Road, Tarnaka, Hyderabad, TS, 500007, India; Academy of Scientific and Innovative Research, Ghaziabad, 201002, India.
| |
Collapse
|
225
|
Zheng X, Zhang X, Yu J, Zheng J. Pan-cancer analysis identifies EIPR1 as a potential prognostic and immunological biomarker for lung adenocarcinoma and its functional validation. Gene 2025; 954:149439. [PMID: 40154585 DOI: 10.1016/j.gene.2025.149439] [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: 03/15/2025] [Accepted: 03/23/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND EARP and GARP complex-interacting protein 1 (EIPR1) may be a new oncogene in tumors, influencing the prognosis and invasion of cancer. However, a systematic analysis of the function of EIPR1 in various cancers remains vacant. Thus, we proceeded with a comprehensive analysis to ascertain the role of EIPR1 among various cancers. METHODS We explored EIPR1 expression in pan-cancer, and its association with clinical stage, survival, gene mutations and methylation by the TIMER 2.0, GEPIA2, cBioPortal, and UALCAN. The protein-protein interaction (PPI) network, immune infiltration, and immune checkpoint assessments of EIPR1 was performed using the STRING and SangerBox. The role of EIPR1 expression in lung adenocarcinoma (LUAD) was explored by the R software. The impact of EIPR1 expression on LUAD progression was studied through in vitro assays. RESULTS EIPR1 was overexpressed in most cancers and revealed as a potential prognostic biomarker in tumors, involving in tumorigenesis by affecting its methylation and gene mutations. The immune infiltration and immune checkpoints of tumors were related to the expression of EIPR1. Additionally, EIPR1 expression affected the survival, diagnosis, clinicopathological features, tumor microenvironment, and drug sensitivity of LUAD patients. Validation studies demonstrated that EIPR1 knockdown suppressed the malignant growth, invasion, and migration of LUAD cells. CONCLUSIONS This study delivers an extensive landscape for the oncogenesis and immunological characteristics of EIPR1, which reveals that EIPR1 may serve as a potential biological target for future prognosis and immune treatment in tumors, especially in LUAD.
Collapse
Affiliation(s)
- Xin Zheng
- Department of Diagnostic Pathology, Shandong Second Medical University, Weifang 261053, China
| | - Xiao Zhang
- Department of Ultrasound, Weifang People's Hospital, Weifang 261041, China
| | - Jie Yu
- Department of Diagnostic Pathology, Shandong Second Medical University, Weifang 261053, China
| | - Jie Zheng
- Department of Diagnostic Pathology, Shandong Second Medical University, Weifang 261053, China; Neurologic Disorders and Regenerative Repair Lab of Shandong Higher Education, Shandong Second Medical University, Weifang 261053, China.
| |
Collapse
|
226
|
Issa H, Singh L, Lai KS, Parusheva-Borsitzky T, Ansari S. Dynamics of inflammatory signals within the tumor microenvironment. World J Exp Med 2025; 15:102285. [DOI: 10.5493/wjem.v15.i2.102285] [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: 10/13/2024] [Revised: 12/31/2024] [Accepted: 01/11/2025] [Indexed: 04/16/2025] Open
Abstract
Tumor stroma, or tumor microenvironment (TME), has been in the spotlight during recent years for its role in tumor development, growth, and metastasis. It consists of a myriad of elements, including tumor-associated macrophages, cancer-associated fibroblasts, a deregulated extracellular matrix, endothelial cells, and vascular vessels. The release of proinflammatory molecules, due to the inflamed microenvironment, such as cytokines and chemokines is found to play a pivotal role in progression of cancer and response to therapy. This review discusses the major key players and important chemical inflammatory signals released in the TME. Furthermore, the latest breakthroughs in cytokine-mediated crosstalk between immune cells and cancer cells have been highlighted. In addition, recent updates on alterations in cytokine signaling between chronic inflammation and malignant TME have also been reviewed.
Collapse
Affiliation(s)
- Hala Issa
- Division of Health Sciences, Higher Colleges of Technology, Abu Dhabi 25026, United Arab Emirates
| | - Lokjan Singh
- Department of Microbiology, Karnali Academy of Health Sciences, Jumla 21200, Karnali, Nepal
| | - Kok-Song Lai
- Division of Health Sciences, Higher Colleges of Technology, Abu Dhabi 25026, United Arab Emirates
| | - Tina Parusheva-Borsitzky
- Division of Health Sciences, Higher Colleges of Technology, Abu Dhabi 25026, United Arab Emirates
| | - Shamshul Ansari
- Division of Health Sciences, Higher Colleges of Technology, Abu Dhabi 25026, United Arab Emirates
| |
Collapse
|
227
|
Liu D, Jiang Y, Ma B, Li L. Structure-based artificial intelligence-aided design of MYC-targeting degradation drugs for cancer therapy. Biochem Biophys Res Commun 2025; 766:151870. [PMID: 40288261 DOI: 10.1016/j.bbrc.2025.151870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
The MYC protein is an oncoprotein that plays a crucial role in various cancers. Although its significance has been well recognized in research, the development of drugs targeting MYC remains relatively slow. In this study, we developed a novel MYC peptide inhibitor based on the MYC/MAX dimer structure, integrating artificial intelligence-assisted peptide drug design. Additionally, we introduced a chaperone-mediated autophagy signal to construct a MYC-targeted degradation drug, MYC-LYSO. By incorporating nano-selenium delivery, we further formulated an enhanced MYC degradation agent, Se-MYC-LYSO. Se-MYC-LYSO demonstrated potent efficacy in inducing MYC degradation, inhibiting tumor cell proliferation, and promoting apoptosis. Moreover, our findings indicate that the efficacy of Se-MYC-LYSO is dependent on the autophagy pathway. These results provide a novel strategy for targeting MYC in cancer therapy.
Collapse
Affiliation(s)
- Donghua Liu
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yize Jiang
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Bohan Ma
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.
| | - Lei Li
- Department of Urology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
228
|
Kim G, Cha Y, Baek SH. Identification of KANK1 as a tumor suppressor gene in pancreatic ductal adenocarcinoma. Biochem Biophys Res Commun 2025; 766:151885. [PMID: 40288262 DOI: 10.1016/j.bbrc.2025.151885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2025] [Accepted: 04/23/2025] [Indexed: 04/29/2025]
Abstract
Pancreatic cancer is a highly lethal malignancy with poor survival outcomes, primarily due to late-stage diagnosis and resistance to conventional therapies. Identifying key oncogenes and tumor suppressor genes is therefore critical for the development of effective treatment strategies. In this study, we identified KANK1 as a novel tumor suppressor gene in pancreatic ductal adenocarcinoma (PDAC) through an integrated mRNA-protein abundance correlation analysis. Elevated KANK1 expression was consistently associated with improved patient survival across multiple datasets, whereas its expression was markedly reduced in pancreatic tumors compared to normal tissues. Single-cell RNA sequencing and immunoblot analyses confirmed the downregulation of KANK1 at both the mRNA and protein levels in PDAC. Further investigation revealed that KANK1 downregulation is driven by copy number loss and tumor hypoxia, supported by data from the TCGA and CCLE databases and validated experimentally under hypoxic conditions. Functional assays demonstrated that KANK1 knockdown promotes pancreatic cancer cell proliferation and migration, along with activation of ERK signaling. Collectively, our findings establish KANK1 as a tumor suppressor in PDAC, whose loss facilitates tumor progression and presents a potential therapeutic target for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Gibeom Kim
- Creative Research Initiatives Center for Epigenetic Code and Diseases, Seoul National University, Seoul, 08826, South Korea; Department of Biological Sciences, Seoul National University, Seoul, 08826, South Korea
| | - Yoonho Cha
- Creative Research Initiatives Center for Epigenetic Code and Diseases, Seoul National University, Seoul, 08826, South Korea; Department of Biological Sciences, Seoul National University, Seoul, 08826, South Korea
| | - Sung Hee Baek
- Creative Research Initiatives Center for Epigenetic Code and Diseases, Seoul National University, Seoul, 08826, South Korea; Department of Biological Sciences, Seoul National University, Seoul, 08826, South Korea.
| |
Collapse
|
229
|
Riviere C, Aljieli M, Mévélec MN, Lantier L, Boursin F, Lajoie L, Ducournau C, Germon S, Moiré N, Dimier-Poisson I, Aubrey N, di Tommaso A. Neospora caninum as delivery vehicle for anti-PD-L1 scFv-Fc: A novel approach for cancer immunotherapy. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200968. [PMID: 40236994 PMCID: PMC11999461 DOI: 10.1016/j.omton.2025.200968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 12/29/2024] [Accepted: 03/16/2025] [Indexed: 04/17/2025]
Abstract
Neospora caninum, a potential anticancer agent able to reactivate the immune response within the tumor microenvironment (TME), has recently shown enhanced immunomodulatory properties in different tumor models when armed with the cytokine, Il-15. In the current area of combination immunotherapy strategies designed to overcome treatment resistance, we engineered for the first time the protozoan Neospora caninum to vectorize and secrete a single-chain variable fragment fused to fragment crystallizable region (scFv-Fc) targeting human programmed cell death ligand 1 (PD-L1). Following validation of its secretion through the micronemes (protozoa secretory organelles), we demonstrated that the scFv-Fc could bind PD-L1 on mouse and human tumor cells, block the programmed cell death protein 1 (PD-1)/PD-L1 pathway leading to potentiate the T cell lymphocyte activity. Additionally, the scFv-Fc induced antibody-dependent cellular phagocytosis (ADCP) and antibody-dependent cellular cytotoxicity (ADCC). Those data demonstrate the feasibility of vectoring and secreting a functional antibody fragment by N. caninum, opening promising avenues for future research.
Collapse
Affiliation(s)
- Clément Riviere
- BioMAP, UMR ISP 1282 INRAe – Université de Tours, 37200 Tours, France
| | - Muna Aljieli
- BioMAP, UMR ISP 1282 INRAe – Université de Tours, 37200 Tours, France
- Faculty of Pharmacy, University of Gezira, Wad Madani, Sudan
| | | | - Louis Lantier
- BioMAP, UMR ISP 1282 INRAe – Université de Tours, 37200 Tours, France
| | - Fanny Boursin
- BioMAP, UMR ISP 1282 INRAe – Université de Tours, 37200 Tours, France
| | - Laurie Lajoie
- BioMAP, UMR ISP 1282 INRAe – Université de Tours, 37200 Tours, France
| | - Céline Ducournau
- BioMAP, UMR ISP 1282 INRAe – Université de Tours, 37200 Tours, France
| | - Stéphanie Germon
- BioMAP, UMR ISP 1282 INRAe – Université de Tours, 37200 Tours, France
| | - Nathalie Moiré
- BioMAP, UMR ISP 1282 INRAe – Université de Tours, 37200 Tours, France
| | | | - Nicolas Aubrey
- BioMAP, UMR ISP 1282 INRAe – Université de Tours, 37200 Tours, France
| | - Anne di Tommaso
- BioMAP, UMR ISP 1282 INRAe – Université de Tours, 37200 Tours, France
| |
Collapse
|
230
|
Wan D, Zhang Q, Yang Z, Zhang X, Xie P, Cheng S, Xu L, Liu B, Zhang K, Zhang W. Engineered oncolytic virus OH2-FLT3L enhances antitumor immunity via dendritic cell activation. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200975. [PMID: 40236993 PMCID: PMC11999459 DOI: 10.1016/j.omton.2025.200975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 02/17/2025] [Accepted: 03/18/2025] [Indexed: 04/17/2025]
Abstract
The combination of oncolytic viruses (OVs) with other immunotherapies, such as immunostimulatory therapies, is a current research hotspot; however, optimizing their therapeutic potential remains to be fully explored. Here, we designed a novel oncolytic herpes simplex virus 2 expressing Fms-like tyrosine kinase 3 ligand (OH2-FLT3L), which induces an antitumor cytotoxic T cell immune response by activating dendritic cells (DCs). We found that OH2-FLT3L specifically infects tumor cells, induces immunogenic cell death (ICD), and releases a large number of tumor-specific antigens, which bound to danger signals and facilitated antigenic cross-presentation by DCs, significantly enhancing T cell activation and function. Experimental results showed that OH2-FLT3L significantly increased the proportion of activated DCs, enhanced the antitumor immune response, and effectively converted "cold" tumors into "hot" tumors. In addition, when combined with anti-PD-1 antibody, OH2-FLT3L further enhanced therapeutic efficacy. In conclusion, OH2-FLT3L, as a novel oncolytic virus, demonstrates the potential to enhance antitumor immune responses through DC activation.
Collapse
Affiliation(s)
- Duo Wan
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Qi Zhang
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Zhenrong Yang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Xiaoli Zhang
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Peipei Xie
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Shujun Cheng
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Libin Xu
- Department of Orthopedic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Binlei Liu
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, College of Bioengineering, Hubei University of Technology, Wuhan 430068, China
| | - Kaitai Zhang
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Wen Zhang
- Department of Immunology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
231
|
Goloudina A, Le Chevalier F, Authié P, Charneau P, Majlessi L. Shared neoantigens for cancer immunotherapy. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200978. [PMID: 40256120 PMCID: PMC12008704 DOI: 10.1016/j.omton.2025.200978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
Exploration of neoantigens holds the potential to be productive in immuno-oncotherapy. Among tumor-specific antigens, neoantigens result from genetic instability that gives rise to non-synonymous somatic mutations, highly specific to tumor cells. In addition to point mutations, gene rearrangements, indels leading to frameshifts, chromosomal translocations or inversions that may lead to fusion proteins, alternative mRNA splicing, and integration of genetic material of oncogenic viruses into the host genome provide consistent sources of neoantigens that are absent in healthy tissues. Out of these alterations, 2%-3% may generate T cell neoepitopes, possibly detectable by TCRs. Neoantigens are absent in healthy tissues and are thus at low risk of triggering autoimmunity. In addition, the host lymphocytes have not been rendered tolerant toward them and it is possible to induce immune responses against them. Here, we overview the two categories of neoantigens, i.e., private and shared, and their use in immuno-oncotherapy in selected pre-clinical and clinical studies. The vast majority of commonly occurring tumor-specific mutations are cancer causing and are permanently expressed by all malignant tumor cells, preventing the latter from escaping vaccine-induced anti-neoantigen immunity. The use of public neoantigens combined with efficient vaccine platforms can provide non-personalized "off-the-shelf" therapeutic vaccine candidates for broad-spectrum immunotherapy purposes.
Collapse
Affiliation(s)
- Anastasia Goloudina
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 rue du Dr. Roux, 75015 Paris, France
| | - Fabien Le Chevalier
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 rue du Dr. Roux, 75015 Paris, France
| | - Pierre Authié
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 rue du Dr. Roux, 75015 Paris, France
| | - Pierre Charneau
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 rue du Dr. Roux, 75015 Paris, France
| | - Laleh Majlessi
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 rue du Dr. Roux, 75015 Paris, France
| |
Collapse
|
232
|
Dutta A, Chakraborty S, Bhattacharya A, Basak U, Pati S, Mukherjee S, Guha D, Banerjee S, Chaudhuri NR, Sarkar D, Jana K, Sa G, Dastidar SG, Das T. hsa-miR-5688 inhibits FOXC1-OCT4/SOX2 feedforward loop that drives chemoresistance in breast cancer stem cells. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200982. [PMID: 40330903 PMCID: PMC12051596 DOI: 10.1016/j.omton.2025.200982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 02/15/2025] [Accepted: 04/04/2025] [Indexed: 05/08/2025]
Abstract
Inherently chemotherapy-resistant breast cancer stem cells (CSCs) are responsible for tumor initiation, metastasis, and relapse. CSCs "acquire" more resistance and stemness upon chemotherapy, thereby making relapse-free survival extremely challenging. Here, we describe a novel role of FOXC1 in "acquired resistance" of breast CSCs during chemotherapy. Putative binding sites of pluripotency factors OCT4 and SOX2, but not NANOG, on FOXC1 promoter, were demonstrated by JASPAR and validated by a docking experiment. Significant decline in FOXC1 expression was noticed after OCT4 or SOX2 ablation in breast CSCs. Contrastingly, presence of putative FOXC1 binding sites on the promoters of stemness genes and drug-resistance marker ABCG2, along with downregulation of OCT4 and SOX2 in FOXC1-ablated CSCs, indicated the existence of a feedforward FOXC1-OCT4/SOX2 transactivation loop in CSCs. Chemotherapy-induced upregulation of FOXC1, stemness, as well as drug resistance in CSCs, and downregulation of the same by prior FOXC1-ablation in in-vitro and in-vivo models, endorsed the contribution of this loop in chemo-induced acquisition of stemness and drug resistance. Finally, over-expression of hsa-miR-5688 sensitized CSCs toward chemotherapy and decelerated recurrence. Accordingly, we demonstrate a hitherto unknown mechanism underpinning chemotherapy-induced resistance in breast CSCs, causing relapse and identified hsa-miR-5688 as a potential therapeutic candidate for relapse-free survival of breast cancer patients.
Collapse
Affiliation(s)
- Apratim Dutta
- Centenary Building, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India
| | - Sourio Chakraborty
- Centenary Building, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India
| | - Apoorva Bhattacharya
- Centenary Building, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India
| | - Udit Basak
- Centenary Building, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India
| | - Subhadip Pati
- Centenary Building, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India
| | - Sumon Mukherjee
- Centenary Building, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India
| | - Deblina Guha
- Centenary Building, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India
| | - Shruti Banerjee
- Centenary Building, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India
| | - Nibedita Ray Chaudhuri
- Unified Academic Campus, Bose Institute, HCGJ+4X5, EN Block, Sector V, Bidhannagar, Kolkata, West Bengal 700091, India
| | - Diptendra Sarkar
- Department of Surgery, IPGMER and SSKM Hospital, Kolkata 700020, India
| | - Kuladip Jana
- Unified Academic Campus, Bose Institute, HCGJ+4X5, EN Block, Sector V, Bidhannagar, Kolkata, West Bengal 700091, India
| | - Gaurisankar Sa
- Centenary Building, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India
| | - Shubhra Ghosh Dastidar
- Unified Academic Campus, Bose Institute, HCGJ+4X5, EN Block, Sector V, Bidhannagar, Kolkata, West Bengal 700091, India
| | - Tanya Das
- Centenary Building, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India
| |
Collapse
|
233
|
Tolstyka ZP, Grohar PJ. Exploiting divergent mechanisms of trabectedin for bone tumors. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200959. [PMID: 40151540 PMCID: PMC11937656 DOI: 10.1016/j.omton.2025.200959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Affiliation(s)
- Zachary P. Tolstyka
- University of Michigan Medical School, Department of Pediatrics, Division of Pediatric Hematology/Oncology, Rogel Cancer Center, 1500 East Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Patrick J. Grohar
- University of Michigan Medical School, Department of Pediatrics, Division of Pediatric Hematology/Oncology, Rogel Cancer Center, 1500 East Medical Center Drive, Ann Arbor, MI 48109, USA
| |
Collapse
|
234
|
Xu S, Yuan X, Wang Y, Fu Z, Chen K, Cui Z, Xu L, Zhang H, Xia D, Wu Y. Bisphenols exposure at environmentally relevant dose promoted ovarian cancer progression and modulated tumor microenvironment through β-catenin/SPP1 axis. JOURNAL OF HAZARDOUS MATERIALS 2025; 490:137824. [PMID: 40054195 DOI: 10.1016/j.jhazmat.2025.137824] [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: 09/15/2024] [Revised: 02/18/2025] [Accepted: 03/01/2025] [Indexed: 04/16/2025]
Abstract
Bisphenol A (BPA) and its substitute, Bisphenol S (BPS) are typical endocrine-disrupting chemicals used in plastics, but their cancer-promoting effect has remained controversial. Here, we investigated the effects of environmentally relevant doses of BPA/BPS exposure on the tumor microenvironment (TME) in ovarian cancer. BPA exposure levels was exhibiting a declining trend and BPS showing an ascending trend in the female population by analyzing the NHANES data (2013-2016). Low doses of BPA/BPS both significantly promoted the migration and invasion of ovarian cancer cells in a dose-dependent manner by activating the Wnt/β-catenin signaling pathway, thereby facilitating the SPP1 gene transcription. Notably, low-dose BPA/BPS exposure stimulated ovarian cancer cells to secrete OPN protein (coded by the SPP1 gene), subsequently inducing the transformation of fibroblasts into cancer-associated fibroblasts (CAFs), which could reshape the TME of ovarian cancer. Two in-vivo experiments established with nude mice and SPP1-/- mice respectively, both confirmed that low-dose BPA/BPS exposure increased the incidence of tumor metastasis accompanied by CAF infiltration, while administration of OPN-neutralizing antibodies effectively blocked these effects. Our results indicated that exposure to either BPA or its substitute BPS could promote the release of secreted protein OPN via the β-catenin/SPP1 axis, ultimately modulating the TME and enhancing the progression of ovarian cancer, providing new evidence and potential intervention strategies for the toxicological assessment and management of bisphenols.
Collapse
Affiliation(s)
- Sinan Xu
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoyu Yuan
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuwei Wang
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhiqin Fu
- Department of Gynecology and Obstetrics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kelie Chen
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Gynecology and Obstetrics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhenyan Cui
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Leting Xu
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Honghe Zhang
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Dajing Xia
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Yihua Wu
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
235
|
Zhang S, Zhou H, Liu Y, Chen G, Li Q, Xu Y, Zheng R, Li S, Chen X, Zhao L. A stimuli-responsive immunostimulant to activate chemo-immunotherapeutic effects by inducing DNA damage and STING activation. J Colloid Interface Sci 2025; 688:664-676. [PMID: 40022787 DOI: 10.1016/j.jcis.2025.02.179] [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: 01/18/2025] [Revised: 02/23/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025]
Abstract
The integration of chemotherapy with systemic immune activation represents a promising approach to eradicate metastatic tumors. In this study, a stimuli-responsive immunostimulant nanoplatform (denoted as MV@Lip) was developed to synergistically activate antitumor immunity via chemotherapy-induced DNA damage and subsequent activation of the stimulator of interferon genes (STING) signaling pathway. The MV@Lip system encapsulates the chemotherapeutic agent mitoxantrone (Mit) and the STING agonist vadimezan (Vad) within a redox-responsive liposomal carrier. MV@Lip was indicated to enhance drug stability, while simultaneously promoting efficient co-delivery of both agents into tumor cells and suppressing tumor proliferation. Mechanistically, MV@Lip exerts its therapeutic efficacy through inducing DNA damage and triggering immunogenic cell death (ICD) to enhance tumor immunogenicity by releasing damage-associated molecular patterns (DAMPs). Concurrently, the released Vadimezan could activate the STING pathway, amplifying innate immune responses through the production of proinflammatory factors and the recruitment of immune effector cells. This concerted action facilitates the infiltration and activation of natural killer (NK) cells and T lymphocytes in the tumor microenvironment, ultimately leading to the suppression of both primary and metastatic tumors. These findings provide a compelling basis for advancing chemotherapeutic combinations as immune-stimulating strategies in the treatment of metastatic malignancies.
Collapse
Affiliation(s)
- Shuiying Zhang
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, PR China
| | - Hangyu Zhou
- The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, the School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Yixin Liu
- The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, the School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Guangmiao Chen
- The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, the School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Qiuyuan Li
- The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, the School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Youqin Xu
- The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, the School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Rongrong Zheng
- The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, the School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Shiying Li
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, PR China; The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, the School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, PR China.
| | - Xin Chen
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, PR China.
| | - Linping Zhao
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, PR China; The Fifth Affiliated Hospital, Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, the School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, PR China.
| |
Collapse
|
236
|
Yin H, Dong X, Liu T. Identification of novel AURKA inhibitors against neuroblastoma using a virtual screening approach. Bioorg Chem 2025; 160:108480. [PMID: 40253760 DOI: 10.1016/j.bioorg.2025.108480] [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: 03/18/2025] [Revised: 04/04/2025] [Accepted: 04/14/2025] [Indexed: 04/22/2025]
Abstract
This study aims to screen and validate AURKA inhibitors to provide lead compounds and theoretical foundations for targeted therapy of neuroblastoma (NB). Through computer-aided drug screening, 11 compounds effectively binding to AURKA were selected from the YaTCM database, and their toxicity was predicted using admetSAR. Subsequently, molecular dynamics (MD) simulations were employed to evaluate the binding affinity and complex stability of the compounds with AURKA, leading to the identification of four preferred compounds (Erylatissin B, (+)-khellactone, Brazilin, and hematoxylin). Further steered molecular dynamics (SMD) and umbrella sampling (US) simulations were conducted to calculate the dissociation energy, confirming their binding strength with AURKA. In vitro experiments demonstrated that Brazilin significantly inhibited proliferation, migration, and induced apoptosis in SK-N-BE (2) cells, while also suppressing AURKA protein expression and its interaction with N-Myc. In vivo experiments showed that Brazilin markedly inhibited tumor growth in a mouse NB model. The findings indicate that Brazilin, by targeting AURKA, exhibits potential anti-NB activity, offering a new candidate compound and theoretical support for NB treatment.
Collapse
Affiliation(s)
- Hongli Yin
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215003, China.
| | - Xue Dong
- Department of Pharmaceutics, Dalian Women and Children's Medical Group, Dalian, Liaoning 116012, China
| | - Tianyi Liu
- Department of Pharmaceutics, Dalian Women and Children's Medical Group, Dalian, Liaoning 116012, China.
| |
Collapse
|
237
|
Sekar Y, Ishwar D, Tan B, Venkatakrishnan K. Nano biosensor unlocks tumor derived immune signals for the early detection of ovarian cancer. Biosens Bioelectron 2025; 278:117368. [PMID: 40088704 DOI: 10.1016/j.bios.2025.117368] [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: 01/08/2025] [Revised: 02/25/2025] [Accepted: 03/10/2025] [Indexed: 03/17/2025]
Abstract
Ovarian cancer is a critical health issue for women nowadays. Its impact is significant because of its high mortality rate (324,603 worldwide), late-stage diagnosis and poor survival rate. Lack of screening tests, vague symptoms, misdiagnosis, and age factor makes it even more difficult to detect. Neutrophils, a subset of immune cells, undergo tumor-specific changes as ovarian cancer progresses inside ovarian tumour microenvironment. Therefore, monitoring the time-specific activity of neutrophils in circulation has the potential to aid in the diagnosis of ovarian cancer. Most ovarian tumor-specific antigens are unknown, making it difficult to identify neutrophils associated with ovarian tumor. We present ovarian tumor-associated circulating neutrophil cell profiling as a stand-alone cancer diagnostic method using a liquid biopsy. Using a SERS-functionalized nano probe, the metabolic profiles of neutrophils from ovarian tumor interaction are detected. We demonstrate that neutrophils associated with cancer stem cells have a distinct metabolic profile and are useful in the diagnosis of early ovarian cancer. Using 5 μL of peripheral blood and an artificial neural network, the characteristics of neutrophil profiles in patient blood could distinguish cancer cohort from non-cancer (healthy) with a 90 % sensitivity and 100 % specificity. Our results demonstrate the viability of using circulating neutrophils for non-invasive cancer diagnostics.
Collapse
Affiliation(s)
- Yuvaraj Sekar
- Institute for Biomedical Engineering, Science and Technology (I BEST), Partnership Between Toronto Metropolitan University and St. Michael's Hospital, Toronto, Ontario, M5B 1W8, Canada; Ultrashort Laser Nanomanufacturing Research Facility, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University, 350 Victoria Street, Toronto, ON, M5B 2K3, Canada; Nano Characterization Laboratory, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University, 350 Victoria Street, Toronto, Ontario, M5B 2K3, Canada; Nano-Bio Interface Facility, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University, 350 Victoria Street, Toronto, ON, M5B 2K3, Canada
| | - Deeptha Ishwar
- Department of Stomatology, Faculty of Dental Medicine, Universite de Montreal, Montreal, QC, H3C 3J7, Canada
| | - Bo Tan
- Institute for Biomedical Engineering, Science and Technology (I BEST), Partnership Between Toronto Metropolitan University and St. Michael's Hospital, Toronto, Ontario, M5B 1W8, Canada; Nano Characterization Laboratory, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University, 350 Victoria Street, Toronto, Ontario, M5B 2K3, Canada; Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, Ontario, M5B 1W8, Canada
| | - Krishnan Venkatakrishnan
- Institute for Biomedical Engineering, Science and Technology (I BEST), Partnership Between Toronto Metropolitan University and St. Michael's Hospital, Toronto, Ontario, M5B 1W8, Canada; Ultrashort Laser Nanomanufacturing Research Facility, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University, 350 Victoria Street, Toronto, ON, M5B 2K3, Canada; Nano-Bio Interface Facility, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University, 350 Victoria Street, Toronto, ON, M5B 2K3, Canada; Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, Ontario, M5B 1W8, Canada.
| |
Collapse
|
238
|
Lin F, Li J, Zhou L, Yi R, Chen Y, He S. Targeting vulnerability in tumor therapy: Dihydroorotate dehydrogenase. Life Sci 2025; 371:123612. [PMID: 40187643 DOI: 10.1016/j.lfs.2025.123612] [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: 01/16/2025] [Revised: 03/20/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Dihydroorotate dehydrogenase (DHODH) is a key enzyme in the de novo pyrimidine biosynthetic pathway and a recognized therapeutic target in various diseases. In oncology research, DHODH has gained increasing importance and become a hot target for various tumor therapy studies. This review highlights three key points: (1) DHODH enables its diverse biological functions through its unique structural features and dominates the regulation of tumor metabolism and cell fate; (2) DHODH activates oncogenic signals, drives metastatic adaptation, and remodels drug resistance networks in tumors, making it a metabolic-signaling dual hub; and (3) DHODH inhibitors have shown significant efficacy in preclinical models of various tumors but face multiple challenges in clinical trials, including drug-related limitations and external constraints. Given these challenges, future research should explore DHODH inhibitors as a foundation for overcoming technological and translational barriers while establishing a systematic framework for the clinical application of DHODH-targeted tumor therapies.
Collapse
Affiliation(s)
- Fu Lin
- Department of Pathology, School of Basic Medicine and Forensic Science, Baotou Medical College, Baotou 014040, China
| | - Jiaxin Li
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Lei Zhou
- Laboratory of Pathogen Biology and Immunology, School of Basic Medicine and Forensic Science, Baotou Medical College, Baotou 014040, China
| | - Rigui Yi
- Department of Pathology, School of Basic Medicine and Forensic Science, Baotou Medical College, Baotou 014040, China
| | - Yingge Chen
- School of Basic Medicine and Forensic Science, Baotou Medical College, Baotou 014040, China
| | - Shuai He
- Department of Pathology, School of Basic Medicine and Forensic Science, Baotou Medical College, Baotou 014040, China.
| |
Collapse
|
239
|
Orellana AMM, Mazucanti CH, Andreotti DZ, de Sá Lima L, Kawamoto EM, Scavone C. Effects of decrease in Klotho protein expression on insulin signaling and levels of proteins related to brain energy metabolism. Eur J Pharmacol 2025; 997:177587. [PMID: 40187598 DOI: 10.1016/j.ejphar.2025.177587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 03/31/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025]
Abstract
Mutations in Klotho have been associated with premature ageing and cognitive dysfunction. Although highly expressed in specific regions of the brain, the actions of Klotho in the central nervous system (CNS) remain largely unknown. Here, we show that animals with a mutated hypomorphic Klotho gene have altered glycaemic regulation, suggesting higher insulin sensitivity. In the CNS, pathways related to insulin intracellular signalling were found to be up-regulated in the hippocampus, with higher activation of protein kinase B and mammalian target of rapamycin and inactivation of the transcription factors forkhead box O (FOXO)-1 and FOXO-3a. In addition, the present study showed that in the hippocampi of wild-type aged mice, where Klotho is naturally downregulated, the levels of some proteins related to energy metabolism and metabolic coupling between neurones and astrocytes, such as monocarboxylate transporter 2 and 4, 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase isoform 3 and lactate dehydrogenase enzymes isoforms A and B were altered. These findings suggest that Klotho plays an essential role in regulating proteins and genes related to metabolic coupling in the brain.
Collapse
Affiliation(s)
- Ana Maria Marques Orellana
- Laboratory of Molecular Neuropharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil; Laboratory of Molecular and Functional Neurobiology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Caio Henrique Mazucanti
- Laboratory of Molecular Neuropharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil; Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, Maryland, USA
| | - Diana Zukas Andreotti
- Laboratory of Molecular and Functional Neurobiology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Larissa de Sá Lima
- Laboratory of Molecular Neuropharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil; Laboratory of Molecular and Functional Neurobiology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Elisa Mitiko Kawamoto
- Laboratory of Molecular and Functional Neurobiology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Cristoforo Scavone
- Laboratory of Molecular Neuropharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
240
|
Li C, Hua C, Chu C, Jiang M, Zhang Q, Zhang Y, Wu L, Liu J, Yang H, Yu XF, Liu J, Geng S, Yang H. A photothermal-responsive multi-enzyme nanoprobe for ROS amplification and glutathione depletion to enhance ferroptosis. Biosens Bioelectron 2025; 278:117384. [PMID: 40121146 DOI: 10.1016/j.bios.2025.117384] [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: 02/17/2025] [Revised: 03/10/2025] [Accepted: 03/15/2025] [Indexed: 03/25/2025]
Abstract
Ferroptosis therapy employs reactive oxygen species (ROS) generated via Fenton or Fenton-like reactions. However, the antioxidant system associated with the tumor microenvironment (TME) exhibits high levels of glutathione (GSH) that significantly restrict the therapeutic efficacy of ferroptosis. In this study, we propose a near-infrared (NIR)-responsive hollow mesoporous manganese dioxide (HM-MnO2) nanoprobe with multi-enzyme-like activity for enhanced ROS generation and GSH depletion that can efficiently promote ferroptosis. The ferroptosis inducer RSL3 is encapsulated within HM-MnO2 with a loading capacity of 67 %, while iron-doped dopamine (Fe-PDA) and cRGD tumor-targeting peptides are conjugated on the surface. The resultant MnO2R@FePDA-cRGD nanocomposite delivers a photothermal conversion efficiency of 39.1 % under 808 nm irradiation, which can effectively trigger structural degradation of the nanoplatform and the rapid release of RSL3. The photothermal effects significantly augment catalytic activity, enabling a multi-enzyme mimicking that includes peroxidase (POD), oxidase (OXD), GSH peroxidase (GPx), and NADH oxidase (NOx) functions, generating significant ROS radicals and an efficient depletion of intracellular GSH. These cascade reactions contribute to an optimal TME for inducing "explosive" ferroptosis with a synergistic inhibition of tumor growth in vitro and in vivo. The proposed strategy represents a potent approach to amplifying ferroptosis through the photothermal-driven rapid release of RSL3 and enhanced multi-enzyme mimetic activities with significant potential in nanomedicine-based cancer therapy.
Collapse
Affiliation(s)
- Chu Li
- Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China; Shenzhen Key Laboratory of Micro/Nano Biosensing, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; School of Stomatology, Zunyi Medical University, Zunyi Guizhou, 563099, China
| | - Chaolei Hua
- Shenzhen Key Laboratory of Micro/Nano Biosensing, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chenchen Chu
- Shenzhen Key Laboratory of Micro/Nano Biosensing, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Mingyang Jiang
- Shenzhen Key Laboratory of Micro/Nano Biosensing, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Qiongdi Zhang
- Shenzhen Key Laboratory of Micro/Nano Biosensing, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yubei Zhang
- Shenzhen Key Laboratory of Micro/Nano Biosensing, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Lie Wu
- Shenzhen Key Laboratory of Micro/Nano Biosensing, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Jian Liu
- Shenzhen Key Laboratory of Micro/Nano Biosensing, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Huijun Yang
- Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China
| | - Xue-Feng Yu
- Shenzhen Key Laboratory of Micro/Nano Biosensing, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Jianguo Liu
- Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China.
| | - Shengyong Geng
- Shenzhen Key Laboratory of Micro/Nano Biosensing, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Hongyu Yang
- Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China.
| |
Collapse
|
241
|
Botcha L, Sehar M, Cheng YC, Zhang SC, Khan Jadoon MS, Chuang PK. Drug repurposing of 6-AZA-UTP and itraconazole reveals novel B3GALT5 inhibitors for pancreatic cancer. Bioorg Chem 2025; 160:108464. [PMID: 40273705 DOI: 10.1016/j.bioorg.2025.108464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 04/26/2025]
Abstract
Pancreatic cancer has a poor prognosis with limited therapeutic options, necessitating novel treatment strategies. While B3GALT5 enzyme overexpression has been reported in pancreatic cancer cases, effective mechanisms to suppress its activity remain unexplored. In this study, we utilized bioinformatics and in silico studies to evaluate the relationship between B3GALT5 enzyme and pancreatic cancer. Through molecular docking analysis, FDA-approved drugs 6-AZA-UTP and itraconazole were identified as potential B3GALT5 enzyme inhibitors. Biological evaluation on MIA PaCa-2 and AsPC-1 pancreatic cancer cell lines demonstrated that both compounds significantly reduced cell viability. Flow cytometry analysis revealed that both drugs effectively suppressed B3GALT5 enzyme activation by decreasing SSEA-3 expression. Furthermore, both compounds exhibited potent anti-tumor effects by inhibiting cell adhesion, colony formation, and migration while inducing apoptosis in pancreatic cancer cells. Notably, both drugs demonstrated favorable ADMET profiles with no carcinogenic or toxic effects. Our investigations revealed that 6-AZA-UTP and itraconazole can effectively suppress B3GALT5 enzyme activity, resulting in tumor suppression and metastasis inhibition. These findings suggest that either 6-AZA-UTP or itraconazole can inhibit B3GALT5 enzyme activity and may serve as promising therapeutic options for pancreatic cancer treatment through drug repurposing strategy.
Collapse
Affiliation(s)
- Lavanya Botcha
- Institute of Biomedical Sciences, National Sun Yat-Sen University Kaohsiung, 804, Taiwan, ROC
| | - Misbah Sehar
- Institute of Biomedical Sciences, National Sun Yat-Sen University Kaohsiung, 804, Taiwan, ROC
| | - Yi-Chi Cheng
- Institute of Biomedical Sciences, National Sun Yat-Sen University Kaohsiung, 804, Taiwan, ROC
| | - Sheng-Cheng Zhang
- Institute of Biomedical Sciences, National Sun Yat-Sen University Kaohsiung, 804, Taiwan, ROC
| | | | - Po-Kai Chuang
- Institute of Biomedical Sciences, National Sun Yat-Sen University Kaohsiung, 804, Taiwan, ROC.
| |
Collapse
|
242
|
Yang J, Friedman R. Inhibition of FLT3-induced signalling in refractory acute myeloid leukaemia. Bioorg Chem 2025; 160:108424. [PMID: 40209351 DOI: 10.1016/j.bioorg.2025.108424] [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: 01/15/2025] [Revised: 03/10/2025] [Accepted: 03/26/2025] [Indexed: 04/12/2025]
Abstract
Mutations in FLT3 make this receptor tyrosine kinase overactive. Such mutations found in ∼30 % of the patients who suffer from acute myeloid leukaemia (AML). FLT3 mediates signalling networks that lead to cell proliferation and survival. FLT3 inhibitors are used to treat AML but patients who are treated with them typically become resistant. Such resistance often emerges through secondary mutations which either restore the activity of FLT3 in the presence of drugs or activate a key player in a signalling network such as NRAS. We had developed AML-specific cell lines resistant to two advanced FLT3 inhibitors: gilteritinib and FF-10101. Resistance in these cell lines proceeds though different mechanisms. In this study, we followed on the efficacy of five FLT3 inhibitors (gilteritinib, FF-10101 and three promising inhibitors that are being developed), two pan-PI3K inhibitors (one of which also inhibits mTOR) and two c-KIT inhibitors in order to examine the significance of different signalling cascades in FLT3+-AML. In addition, we used molecular modelling and quantum chemistry calculations to explain why specific FLT3 mutations affect some inhibitors more than others. Two novel FLT3 inhibitors were found to be only weakly affected by resistance mutations against gilteritinib and FF-10101. The efficacy of most FLT3 inhibitors was only weakly (or not at all) affected by the NRAS/G12C activating mutation. Finally, no non-FLT3 inhibitor has shown sufficient efficacy in the cells, suggesting the central role of FLT3 in FLT3-mutated AML.
Collapse
MESH Headings
- fms-Like Tyrosine Kinase 3/antagonists & inhibitors
- fms-Like Tyrosine Kinase 3/metabolism
- fms-Like Tyrosine Kinase 3/genetics
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Signal Transduction/drug effects
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/chemistry
- Protein Kinase Inhibitors/chemical synthesis
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/chemical synthesis
- Dose-Response Relationship, Drug
- Molecular Structure
- Structure-Activity Relationship
- Cell Proliferation/drug effects
- Drug Screening Assays, Antitumor
- Drug Resistance, Neoplasm/drug effects
- Cell Line, Tumor
- Aniline Compounds/pharmacology
- Aniline Compounds/chemistry
- Pyrazines/pharmacology
- Pyrazines/chemistry
Collapse
Affiliation(s)
- Jingmei Yang
- Department of Chemistry and Biomedical Science, Linnaeus University, Kalmar SE-39231, Sweden
| | - Ran Friedman
- Department of Chemistry and Biomedical Science, Linnaeus University, Kalmar SE-39231, Sweden.
| |
Collapse
|
243
|
Znaidi R, Massiani-Beaudoin O, Mailly P, Monnet H, Bonnifet T, Joshi RL, Fuchs J. Nuclear translocation of the LINE-1 encoded ORF1 protein alters nuclear envelope integrity in human neurons. Brain Res 2025; 1857:149579. [PMID: 40157412 DOI: 10.1016/j.brainres.2025.149579] [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: 03/06/2025] [Accepted: 03/17/2025] [Indexed: 04/01/2025]
Abstract
LINE-1 retrotransposons are increasingly implicated in aging and neurodegenerative diseases, yet the precise pathogenic mechanisms remain elusive. While the endonuclease and reverse transcriptase activities of LINE-1-encoded ORF2p can induce DNA damage and inflammation, a role of LINE-1 ORF1p in cellular dysfunctions stays unassigned. Here we demonstrate, using a neuronal cellular model, that ORF1p translocates into the nucleus upon arsenite-induced stress, directly interacting with nuclear import (KPNB1), nuclear pore complex (NUP153), and nuclear lamina (Lamin B1) proteins. Nuclear translocation of ORF1p disrupts nuclear integrity, nucleocytoplasmic transport, and heterochromatin structure, features linked to neurodegeneration and aging. Elevated nuclear ORF1p levels induced either by arsenite-induced stress, ORF1p overexpression, or as observed in Parkinson's disease post-mortem brain tissues correlate with impaired nuclear envelope (NE) morphology. Stress-induced nuclear alterations are mitigated by blocking ORF1p nuclear import or with the anti-aging drug remodelin. This study thus reveals a pathogenic action of nuclear ORF1p in human neurons driving NE alterations and thereby contributing to LINE-1-mediated cell toxicity.
Collapse
Affiliation(s)
- Rania Znaidi
- CIRB, Collège de France, Université PSL, CNRS, INSERM, 75005 Paris, France
| | | | - Philippe Mailly
- Orion Imaging Facility, CIRB, Collège de France, Université PSL, CNRS, INSERM, Labex Memolife, 75005 Paris, France
| | - Héloïse Monnet
- Orion Imaging Facility, CIRB, Collège de France, Université PSL, CNRS, INSERM, Labex Memolife, 75005 Paris, France
| | - Tom Bonnifet
- CIRB, Collège de France, Université PSL, CNRS, INSERM, 75005 Paris, France
| | - Rajiv L Joshi
- CIRB, Collège de France, Université PSL, CNRS, INSERM, 75005 Paris, France.
| | - Julia Fuchs
- CIRB, Collège de France, Université PSL, CNRS, INSERM, 75005 Paris, France.
| |
Collapse
|
244
|
Xie J, Shu X, Xie Z, Tang J, Wang G. Pharmacological modulation of cellular senescence: Implications for breast cancer progression and therapeutic strategies. Eur J Pharmacol 2025; 997:177475. [PMID: 40049574 DOI: 10.1016/j.ejphar.2025.177475] [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: 02/26/2025] [Accepted: 03/04/2025] [Indexed: 05/02/2025]
Abstract
Senescence, defined by the cessation of cell proliferation, plays a critical and multifaceted role in breast cancer progression and treatment. Senescent cells produce senescence-associated secretory phenotypes (SASP) comprising inflammatory cytokines, chemokines, and small molecules, which actively shape the tumor microenvironment, influencing cancer development, progression, and metastasis. This review provides a comprehensive analysis of the types and origins of senescent cells in breast cancer, alongside their markers and detection methods. Special focus is placed on pharmacological strategies targeting senescence, including drugs that induce or inhibit senescence, their molecular mechanisms, and their roles in therapeutic outcomes when combined with chemotherapy and radiotherapy. By exploring these pharmacological interventions and their impact on breast cancer treatment, this review underscores the potential of senescence-targeting therapies to revolutionize breast cancer management.
Collapse
Affiliation(s)
- Jialing Xie
- Department of Clinical Pharmacology, Xiangya Hospital, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, People's Republic of China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Changsha, 410008, People's Republic of China
| | - Xianlong Shu
- Department of Clinical Pharmacology, Xiangya Hospital, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, People's Republic of China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Changsha, 410008, People's Republic of China
| | - Zilan Xie
- Department of Clinical Pharmacology, Xiangya Hospital, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, People's Republic of China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Changsha, 410008, People's Republic of China
| | - Jie Tang
- Department of Clinical Pharmacology, Xiangya Hospital, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, People's Republic of China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Changsha, 410008, People's Republic of China.
| | - Guo Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, People's Republic of China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Changsha, 410008, People's Republic of China.
| |
Collapse
|
245
|
Wang P, Ding W, Mo J, Gu C, Ouyang S, Peng K, Zhang Q, Liu G, Lu J, Wang Y, Hu W, Zhu K, Zhang X. A novel adenosine 2A receptor antagonist HZ-086 enhances the efficiency of immunotherapy and alleviates the acquired resistance to PD-L1 by restoration of T cell functions. Eur J Pharmacol 2025; 997:177535. [PMID: 40118325 DOI: 10.1016/j.ejphar.2025.177535] [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: 11/02/2024] [Revised: 03/17/2025] [Accepted: 03/19/2025] [Indexed: 03/23/2025]
Abstract
Immunotherapy faces significant challenges due to low clinical response rates and immune escape mechanisms, which ultimately lead to drug resistance. Previous studies suggest that adenosine-2A receptor (A2AR) signaling plays a critical role in immunosuppression and immune escape. However, no potent and selective A2AR inhibitors are currently available for clinical use to address immunotherapy resistance in tumors. In this study, we identified a novel small molecule compound, HZ-086, as a potent and selective inhibitor of A2AR. HZ-086 restored the activation of T-cell signaling which is suppressed by adenosine analogs in vitro. Additionally, HZ-086 enhanced T-cell-mediated cytotoxicity, increased the secretion of cytokines for antitumor and subsequently inhibited growth of tumor cells in vitro and in vivo. Furthermore, HZ-086 inhibited tumor growth, enhances anti-tumor capacity, and reversed PD-L1 resistance in vivo. When combined with FD-L1, a PD-L1 small molecule inhibitor discovered by our lab, HZ-086 achieved over 80 % tumor growth inhibition (TGI) and restored immune response in anti-PD-L1 monoclonal antibody-resistant tumors. This combination treatment also promoted the infiltration and activation of CD8+ T lymphocytes within the tumor microenvironment. Our findings demonstrate that adenosine-A2AR signaling mediates resistance to immunotherapy and discover a novel potent and selective A2AR inhibitor with high efficacy in enhancing antitumor immune responses and reversing PD-L1 resistance. The combination of A2AR inhibitor and PD-L1 inhibitor represents a promising therapeutic strategy for antitumor therapy.
Collapse
Affiliation(s)
- Pengyan Wang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China; Innovation Practice Center, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Wen Ding
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Jianshan Mo
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Chenxi Gu
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Shumin Ouyang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Keren Peng
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Qiyi Zhang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Guopin Liu
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China; State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Jinjian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Yandong Wang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China; State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Wenhao Hu
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Kai Zhu
- Innovation Practice Center, Changchun University of Chinese Medicine, Changchun, 130117, China.
| | - Xiaolei Zhang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| |
Collapse
|
246
|
Kalter N, Gulati S, Rosenberg M, Ayaz Q, Nguyen J, Wang S, Schroeder B, Li CY, Hendel A. Precise measurement of CRISPR genome editing outcomes through single-cell DNA sequencing. Mol Ther Methods Clin Dev 2025; 33:101449. [PMID: 40225018 PMCID: PMC11987616 DOI: 10.1016/j.omtm.2025.101449] [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: 10/20/2024] [Accepted: 03/11/2025] [Indexed: 04/15/2025]
Abstract
Gene therapy for clinical applications necessitates a comprehensive, accurate, and precise measurement of gene-edited drug products. State-of-the-art pipelines for evaluating editing outcomes rely primarily on bulk sequencing approaches, which are limited to population-level assessment. Here, we leveraged Tapestri, a single-cell sequencing technology for an in-depth analysis of editing outcomes. Using this platform, we characterized the genotype of triple-edited cells simultaneously at more than 100 loci, including editing zygosity, structural variations, and cell clonality. Our findings revealed a unique editing pattern in nearly every edited cell, highlighting the importance of single-cell resolution measurement to ensure the highest safety standards.
Collapse
Affiliation(s)
- Nechama Kalter
- The Institute for Advanced Materials and Nanotechnology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 529002, Israel
| | - Saurabh Gulati
- Mission Bio, 400 E Jamie Ct, Suite 100, South San Francisco, CA 94080, USA
| | - Michael Rosenberg
- The Institute for Advanced Materials and Nanotechnology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 529002, Israel
| | - Qawer Ayaz
- Mission Bio, 400 E Jamie Ct, Suite 100, South San Francisco, CA 94080, USA
| | - Joanne Nguyen
- Mission Bio, 400 E Jamie Ct, Suite 100, South San Francisco, CA 94080, USA
| | - Shu Wang
- Mission Bio, 400 E Jamie Ct, Suite 100, South San Francisco, CA 94080, USA
| | - Benjamin Schroeder
- Mission Bio, 400 E Jamie Ct, Suite 100, South San Francisco, CA 94080, USA
| | - Chieh-Yuan Li
- Mission Bio, 400 E Jamie Ct, Suite 100, South San Francisco, CA 94080, USA
| | - Ayal Hendel
- The Institute for Advanced Materials and Nanotechnology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 529002, Israel
| |
Collapse
|
247
|
Qu S, Dai H. Conjugated STING agonists. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102530. [PMID: 40291379 PMCID: PMC12032345 DOI: 10.1016/j.omtn.2025.102530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
An innate immune system is the first line of defense and prevents the host from infection and attacks the invading pathogens. Stimulator of interferon genes (STING) plays a vital role in the innate immune system. STING activation by STING agonists leads to phosphorylation of TANK-binding kinase 1 (TBK1) and interferon regulatory factor 3 (IRF3) with the release of type I interferons and proinflammatory cytokines, further promoting the adaptive immune response and activating T cells by increased antigen presentation. Natural STING agonist cyclic dinucleotides (CDNs) encounter many defects such as high polarity by negative charges, low stability and circulative half-life, off-target systemic toxicity, and low response efficacy in clinical trials. To overcome these challenges, massive efforts have addressed chemical modifications of CDNs, development of non-CDN STING agonists, and delivery of these STING agonists either by conjugation or liposomes/nanoparticles. Considering there have been a great number of reports regarding nanosystem-aided delivery, here, we examine the development of STING agonists, especially for non-CDNs and their delivery specifically by conjugation strategy, with a focus on the STING agonists in clinical trials and current challenges of their potential in cancer immunotherapy.
Collapse
Affiliation(s)
- Shuhao Qu
- School of Veterinary Medicine, Henan University of Animal Husbandry and Economy, Zhengzhou 450046, China
| | - Hong Dai
- Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| |
Collapse
|
248
|
Huang M, Zou J, Luo B, Sun Y, Yang Z, Kong H, Long X, Sun X, Yang M, Wang X, Liu X, Zhao X. p14 ARF interacts with γ-H2AX and is involved in the DNA damage response. Biochem Biophys Res Commun 2025; 765:151847. [PMID: 40267841 DOI: 10.1016/j.bbrc.2025.151847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2025] [Accepted: 04/16/2025] [Indexed: 04/25/2025]
Abstract
p14ARF(ARF) is a tumor suppressor and functionally related to p53. Emerging evidences suggest that ARF triggers DNA damage in a p53-independent manner. However, it remains to be determined how ARF is involved in DNA damage response. Here, we report that ARF is critical in regulating the formation of DNA damage induced γ-H2AX foci. ARF binds to H2AX through its N-terminal domains to promote the phosphorylation of H2AX. The localization of ARF to the site of DNA breaks facilitates the formation of γ-H2AX foci in response to DNA damage. The knocking down of ARF significantly reduced γ-H2AX production and the number of γ-H2AX foci, leading to increased sensitivity to doxorubicin-induced cell death. Together, we propose that ARF plays a crucial role in DNA damage response through its association with H2AX and regulating γ-H2AX formation.
Collapse
Affiliation(s)
- Minyi Huang
- Clinical Research Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, Shenzhen Key Laboratory of Bone Tissue Repair and Translational Research, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China; Laboratory of Cell Fate and Metabolic Regulation, School of Medicine, Sun Yat-sen University, Shenzhen, 518107, China
| | - Juan Zou
- Clinical Research Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, Shenzhen Key Laboratory of Bone Tissue Repair and Translational Research, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China; Laboratory of Cell Fate and Metabolic Regulation, School of Medicine, Sun Yat-sen University, Shenzhen, 518107, China; International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen, China
| | - Biwei Luo
- Division of Hepatobiliary and Pancreas Surgery, Department of General Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Yanxi Sun
- Laboratory of Cell Fate and Metabolic Regulation, School of Medicine, Sun Yat-sen University, Shenzhen, 518107, China
| | - Zhongzhou Yang
- Clinical Research Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, Shenzhen Key Laboratory of Bone Tissue Repair and Translational Research, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
| | - Huimin Kong
- Clinical Research Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, Shenzhen Key Laboratory of Bone Tissue Repair and Translational Research, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
| | - Xinxu Long
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen, China
| | - Xijun Sun
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen, China
| | - Mo Yang
- Clinical Research Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, Shenzhen Key Laboratory of Bone Tissue Repair and Translational Research, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China.
| | - Xingwu Wang
- Laboratory of Cell Fate and Metabolic Regulation, School of Medicine, Sun Yat-sen University, Shenzhen, 518107, China.
| | - Xiangyu Liu
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen, China.
| | - Xiaocheng Zhao
- Clinical Research Center, Guangdong Provincial Key Laboratory of Digestive Cancer Research, Shenzhen Key Laboratory of Bone Tissue Repair and Translational Research, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China.
| |
Collapse
|
249
|
Qian J, Meng H, Wang Z, Sun Y, Xu X, Shi H, Wang C, Zhou L, Lv X, Yang Y, Gu C. GART promotes multiple myeloma malignancy via tumor stemness mediated by activating the HSP90α/CDK6/β-catenin axis. Eur J Pharmacol 2025; 996:177584. [PMID: 40185325 DOI: 10.1016/j.ejphar.2025.177584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 03/31/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Tumor progression involves the acquisition of progenitor and stem cell-like characteristics. Glycinamide ribonucleotide transformylase (GART) has been studied in solid tumors for its role in promoting cancer cell stemness, but its function in multiple myeloma (MM) remains unclear. This study aims to determine the impact of GART on MM cell proliferation and its potential as a therapeutic target. Elevated GART was associated with MM stem cell-like markers and unfavorable outcomes in MM patients. In vitro experiments using lentivirus-based overexpression and siRNA-mediated knockdown methods demonstrated that GART promoted MM cell proliferation, colony formation, and cell cycle progression. In vivo studies using a chemically induced plasmacytoma mouse model confirmed that GART accelerated MM malignancy. Mechanistic studies revealed that GART binds to and stabilizes the HSP90α protein, thereby upregulating its client protein CDK6. Additionally, GART activated the Wnt/β-catenin pathway, promoting cell proliferation and the expression of stemness genes in MM. Furthermore, a GART inhibitor, pemetrexed (PEM), effectively suppressed cell proliferation and tumor growth in a human cell line-derived xenograft model (CDX). In conclusion, our findings demonstrate that GART promotes MM cell proliferation and tumor stemness by activating the HSP90α/CDK6/β-catenin axis. Targeting GART may be a promising strategy for developing and improving MM treatments.
Collapse
Affiliation(s)
- Jinjun Qian
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, China; School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Han Meng
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ze Wang
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yi Sun
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xiaoning Xu
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Hui Shi
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Cheng Wang
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Lianxin Zhou
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xinyu Lv
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ye Yang
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Chunyan Gu
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, China; School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
250
|
Han D, Yu Z, Zhang K, Gai C, Zhang P, Chai X, Zhuo X, Zhao Q, Zou Y, Zhu L. Design, synthesis, and antitumor activity of stapled peptide inhibitors targeting the RAS-RAF interactions. Eur J Med Chem 2025; 290:117568. [PMID: 40153928 DOI: 10.1016/j.ejmech.2025.117568] [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: 01/03/2025] [Revised: 03/12/2025] [Accepted: 03/23/2025] [Indexed: 04/01/2025]
Abstract
RAS-RAF interactions play a vital role in the RAS-RAF-MEK-ERK signaling pathway, significantly regulating cell proliferation, differentiation, and survival. Some small molecule inhibitors targeting various components of this pathway, such as MRTX849 and AMG 510, have been introduced for clinical application. However, peptide-based drugs encounter several challenges, such as poor cell permeability, low biological stability, and rapid in vivo clearance, which hinder their application. Herein, based on co-crystal complex structures and RAS-RAF interaction hotspots, we identified four linear peptides-Raf-0 to Raf-2 and CRD-0-derived from the α-helical regions of the RAS-binding domain (RBD) and the cysteine-rich domain (CRD) of CRAF. Raf-1 was selected for further modification using a hydrocarbon stapling strategy, capping it with stearic acid at the N-terminal due to its highest binding affinity in the SPR assay. As a result, Sraf-2-1 and Sraf-7-1 bound to KRASG12C with Kd values of 3.56 μM and 2.62 μM, respectively, demonstrating robust anticancer activity in the CCK8 assay. Additionally, Sraf-2-1 and Sraf-7-1 reduced AKT phosphorylation, induced cancer cell apoptosis in a concentration-dependent manner, and effectively inhibited cancer cell migration, showing improved α-helix stability and cell permeability. In summary, our findings indicate that the hydrocarbon stapling strategy and stearic acid tagging enhanced the therapeutic potential of peptide inhibitors, offering methods for targeting RAS in cancer therapy.
Collapse
Affiliation(s)
- Dan Han
- School of Health Sciences and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, PR China; Department of Burn Plastic Surgery, The Second Affiliated Hospital of Second Military Medical University, Shanghai, 200003, PR China
| | - Zhou Yu
- School of Pharmacy, Second Military Medical University, Shanghai, 200433, PR China
| | - Kai Zhang
- School of Pharmacy, Second Military Medical University, Shanghai, 200433, PR China
| | - Conghao Gai
- School of Pharmacy, Second Military Medical University, Shanghai, 200433, PR China
| | - Peichao Zhang
- School of Pharmacy, Second Military Medical University, Shanghai, 200433, PR China
| | - Xiaoyun Chai
- School of Pharmacy, Second Military Medical University, Shanghai, 200433, PR China
| | - Xiaobing Zhuo
- School of Pharmacy, Second Military Medical University, Shanghai, 200433, PR China
| | - Qingjie Zhao
- School of Pharmacy, Second Military Medical University, Shanghai, 200433, PR China.
| | - Yan Zou
- School of Pharmacy, Second Military Medical University, Shanghai, 200433, PR China.
| | - Lie Zhu
- School of Health Sciences and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, PR China; Department of Burn Plastic Surgery, The Second Affiliated Hospital of Second Military Medical University, Shanghai, 200003, PR China.
| |
Collapse
|